151
|
Bao C, Ma Q, Ying X, Wang F, Hou Y, Wang D, Zhu L, Huang J, He C. Histone lactylation in macrophage biology and disease: from plasticity regulation to therapeutic implications. EBioMedicine 2025; 111:105502. [PMID: 39662177 PMCID: PMC11697715 DOI: 10.1016/j.ebiom.2024.105502] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/10/2024] [Accepted: 12/02/2024] [Indexed: 12/13/2024] Open
Abstract
Epigenetic modifications have been identified as critical molecular determinants influencing macrophage plasticity and heterogeneity. Among these, histone lactylation is a recently discovered epigenetic modification. Research examining the effects of histone lactylation on macrophage activation and polarization has grown substantially in recent years. Evidence increasingly suggests that lactate-mediated changes in histone lactylation levels within macrophages can modulate gene transcription, thereby contributing to the pathogenesis of various diseases. This review provides a comprehensive analysis of the role of histone lactylation in macrophage activation, exploring its discovery, effects, and association with macrophage diversity and phenotypic variability. Moreover, it highlights the impact of alterations in macrophage histone lactylation in diverse pathological contexts, such as inflammation, tumorigenesis, neurological disorders, and other complex conditions, and demonstrates the therapeutic potential of drugs targeting these epigenetic modifications. This mechanistic understanding provides insights into the underlying disease mechanisms and opens new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Chuncha Bao
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, PR China
| | - Qing Ma
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University /West China School of Nursing, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xihong Ying
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University /West China School of Nursing, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Fengsheng Wang
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, PR China
| | - Yue Hou
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, PR China
| | - Dun Wang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, PR China
| | - Linsen Zhu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, PR China
| | - Jiapeng Huang
- Clinical Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, PR China.
| | - Chengqi He
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, PR China.
| |
Collapse
|
152
|
Wen P, Zhuo X, Wang L. Skin barrier dysfunction in cutaneous T-cell lymphoma: From pathogenic mechanism of barrier damage to treatment. Crit Rev Oncol Hematol 2025; 205:104559. [PMID: 39549893 DOI: 10.1016/j.critrevonc.2024.104559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 10/30/2024] [Accepted: 11/08/2024] [Indexed: 11/18/2024] Open
Abstract
Cutaneous T-cell lymphoma (CTCL) is a group of non-Hodgkin lymphomas characterized by multiple erythematous patches, plaques, or even nodules on the skin. As the disease progresses, patients develop widespread pruritic skin lesions, leading to skin barrier dysfunction, which significantly impacts their quality of life, appearance, and social adaptation. The pathogenesis of CTCL is not fully understood. Recent studies have recognized the important role of skin barrier dysfunction in the development and progression of CTCL, yet a comprehensive review on this topic is lacking. This review summarizes recent findings on skin barrier dysfunction in CTCL, focusing on physical barrier dysfunction, chronic inflammation, and immune dysregulation. We also discuss current and potential therapies aimed at restoring barrier function in CTCL. By emphasizing the integration of barrier-centric approaches into CTCL management, this review provides valuable insights for improving treatment outcomes.
Collapse
Affiliation(s)
- Pengfei Wen
- Department of Dermatology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan 610041, China.
| | - Xiaoxue Zhuo
- Department of Dermatology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan 610041, China.
| | - Lin Wang
- Department of Dermatology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu, Sichuan 610041, China.
| |
Collapse
|
153
|
Wu T, Ji M, Li T, Luo L. The molecular and metabolic landscape of ferroptosis in respiratory diseases: Pharmacological aspects. J Pharm Anal 2025; 15:101050. [PMID: 40034685 PMCID: PMC11873008 DOI: 10.1016/j.jpha.2024.101050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 03/05/2025] Open
Abstract
Ferroptosis is a form of cell death that occurs when there is an excess of reactive oxygen species (ROS), lipid peroxidation, and iron accumulation. The precise regulation of metabolic pathways, including iron, lipid, and amino acid metabolism, is crucial for cell survival. This type of cell death, which is associated with oxidative stress, is controlled by a complex network of signaling molecules and pathways. It is also implicated in various respiratory diseases such as asthma, chronic obstructive pulmonary disease (COPD), acute lung injury (ALI), lung cancer, pulmonary fibrosis (PF), and the coronavirus disease 2019 (COVID-19). To combat drug resistance, it is important to identify appropriate biological markers and treatment targets, as well as intervene in respiratory disorders to either induce or prevent ferroptosis. The focus is on the role of ferroptosis in the development of respiratory diseases and the potential of targeting ferroptosis for prevention and treatment. The review also explores the interaction between immune cell ferroptosis and inflammatory mediators in respiratory diseases, aiming to provide more effective strategies for managing cellular ferroptosis and respiratory disorders.
Collapse
Affiliation(s)
- Tong Wu
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Miaorong Ji
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| |
Collapse
|
154
|
Chen S, Song X, Lv C. Macrophages and Pulmonary Fibrosis. Curr Mol Med 2025; 25:416-430. [PMID: 39779550 DOI: 10.2174/0115665240286046240112112310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 12/27/2023] [Accepted: 01/04/2024] [Indexed: 01/11/2025]
Abstract
Most chronic respiratory diseases often lead to the clinical manifestation of pulmonary fibrosis. Inflammation and immune disorders are widely recognized as primary contributors to the onset of pulmonary fibrosis. Given that macrophages are predominantly responsible for inflammation and immune disorders, in this review, we first focused on the role of different subpopulations of macrophages in the lung and discussed the crosstalk between macrophages and other immune cells, such as neutrophils, regulatory T cells, NKT cells, and B lymphocytes during pulmonary fibrogenesis. Subsequently, we analyzed the interaction between macrophages and fibroblasts as a possible new research direction. Finally, we proposed that exosomes, which function as a means of communication between macrophages and target cells to maintain cellular homeostasis, are a strategy for targeting lung drugs in the future. By comprehending the mechanisms underlying the interplay between macrophages and other lung cells, we aim to enhance our understanding of pulmonary fibrosis, leading to improved diagnostics, preventative measures, and the potential development of macrophage-based therapeutics.
Collapse
Affiliation(s)
- Shengjun Chen
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou 256603, China
| | - Xiaodong Song
- Department of Cellular and Genetic Medicine, Binzhou Medical University, Yantai 264003, China
| | - Changjun Lv
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou 256603, China
| |
Collapse
|
155
|
Zhang H, Ma G, Lv H, Peng Y. Bibliometric Analysis of Non-coding RNAs and Ischemic Stroke: Trends, Frontiers, and Challenges. Mol Biotechnol 2025; 67:1-15. [PMID: 38064146 DOI: 10.1007/s12033-023-00981-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 10/31/2023] [Indexed: 01/03/2025]
Abstract
More and more articles have shown that non-coding RNAs (ncRNAs) play a significant role in the pathogenesis and prognosis of ischemic stroke. However, the bibliometric analysis in ncRNAs and ischemic stroke is still lacking. This study retrieved the Web of Science Core Collection for relevant articles from January 1, 2010 to April 6, 2023. Bibliometrix R, VOSviewer, and CiteSpace were used to perform the bibliometric analysis. A total of 1058 articles were eligible for this review. The number of publications showed a fluctuating upward trend. The total citations were 28,698 times, and the average number of citations per article was 27.12 times. Our findings indicated ncRNAs has been increasingly investigated for its critical role in apoptosis, autophagy, angiogenesis, inflammation, oxidative stress, and blood-brain barrier after ischemic stroke by regulating target mRNAs, extracellular secretion, target proteins, and others. The microRNAs, circular RNAs, and long ncRNAs may be hotspots, and ferroptosis, METTL3, and exosome might be frontier in this field. Besides, ncRNAs have a promising future as diagnostic and prognostic biomarkers, molecular drug targets, and other targeted therapies for ischemic stroke. However, it still faces many challenges to be successfully applied in the clinical practice.
Collapse
Affiliation(s)
- Hanrui Zhang
- Department of Acupuncture and Rehabilitation, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Guquan Ma
- Department of Acupuncture and Rehabilitation, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Hequn Lv
- Department of Acupuncture and Rehabilitation, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Yongjun Peng
- Department of Acupuncture and Rehabilitation, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| |
Collapse
|
156
|
Li K, Liu X, Lu R, Zhao P, Tian Y, Li J. Bleomycin pollution and lung health: The therapeutic potential of peimine in bleomycin-induced pulmonary fibrosis by inhibiting glycolysis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 289:117451. [PMID: 39626488 DOI: 10.1016/j.ecoenv.2024.117451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 11/25/2024] [Accepted: 11/29/2024] [Indexed: 01/26/2025]
Abstract
The increasing use of anticancer drugs has led to the emergence of environmental contaminants such as bleomycin (BLM), which poses significant threats to both aquatic ecosystems and human health. Bleomycin, known for its DNA-damaging properties, is extensively used in oncology. Its resistance to biodegradation, along with the limitations of conventional wastewater treatment processes, facilitates environmental accumulation from various sources, highlighting the need for effective management and treatment strategies to mitigate ecological and health risks. This study investigates the link between BLM pollution and pulmonary fibrosis, a progressive lung disease characterized by tissue scarring and loss of function. We demonstrate that BLM induces pulmonary fibrosis in mice and enhances glycolysis and fibroblast activation. Our findings also indicate that peimine, a natural compound derived from Fritillaria, suppresses fibroblast activation and ameliorates pulmonary fibrosis by inhibiting glycolysis through the PI3K/Akt/PFKFB3 signaling pathway. Taken together, this study underscores the environmental and health risks associated with the accumulation of cytostatic drugs like BLM and highlights the therapeutic potential of natural compounds such as peimine. Our results contribute to the development of novel strategies for the prevention and treatment of pulmonary fibrosis and call for better management practices to mitigate the environmental impact of cytostatic drugs.
Collapse
Affiliation(s)
- Kangchen Li
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Xuefang Liu
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Ruilong Lu
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Peng Zhao
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Yange Tian
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450000, China.
| | - Jiansheng Li
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450000, China; Department of Respiratory Diseases, the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China.
| |
Collapse
|
157
|
Akhlaghpasand M, Tavanaei R, Hosseinpoor M, Heidari R, Mohammadi I, Chamanara M, Hosseinpour M, Zali A, Mosaed R, Oraee-Yazdani S. Effects of Combined Intrathecal Mesenchymal Stem Cells and Schwann Cells Transplantation on Neuropathic Pain in Complete Spinal Cord Injury: A Phase II Randomized Active-Controlled Trial. Cell Transplant 2025; 34:9636897241298128. [PMID: 39874104 PMCID: PMC11775971 DOI: 10.1177/09636897241298128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/09/2024] [Accepted: 10/23/2024] [Indexed: 01/30/2025] Open
Abstract
Neuropathic pain is a debilitating complication following spinal cord injury (SCI). Currently, effective treatments for SCI-induced neuropathic pain are highly lacking. This clinical trial aimed to investigate the efficacy of combined intrathecal injection of Schwann cells (SCs) and bone marrow-derived mesenchymal stem cells (BMSCs) in improving SCI-induced neuropathic pain. This study was a parallel-group, randomized, open-label, active-controlled phase II trial with two arms, including treatment and control groups. Patients with complete SCI-induced neuropathic pain in the treatment group received a single combined intrathecal injection of BMSCs and SCs. Study outcome measures were International SCI Pain Basic Data Set (ISCIPBDS) and World Health Organization (WHO) Quality of Life Assessment Instrument (WHOQOL-BREF). A total of 37 (55.2%) and 30 (44.8%) patients in the treatment and control groups were followed up for 6 months, respectively. Significant reductions in mean scores of interference items in the treatment group, including daily activities (P < 0.001), mood (P < 0.001), and sleep (P < 0.001), were found at 6 months after the injection compared with the control one. Similarly, pain frequency (P = 0.002), mean (P = 0.001), and worst (P = 0.001) numeric rating scale (NRS) pain intensity scores showed significant reductions in the treatment group after 6 months compared with the control one. Based on multiple regression analysis controlled for potential confounders, significant associations between changes in all outcome measures over the study period and the treatment group were found. This clinical trial indicated the efficacy of combined cell therapy in improving the neuropathic pain and quality of life in complete SCI patients. Future investigations should evaluate the effects of combination of this strategy with other existing therapies for SCI-induced neuropathic pain. This clinical trial was also registered prospectively at the Iranian Registry of Clinical Trials (IRCT20200502047277N8).
Collapse
Affiliation(s)
| | - Roozbeh Tavanaei
- Cancer Epidemiology Research Center, AJA University of Medical Sciences, Tehran, Iran
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maede Hosseinpoor
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Heidari
- Cancer Epidemiology Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Ida Mohammadi
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Chamanara
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Melika Hosseinpour
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Zali
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Mosaed
- Cancer Epidemiology Research Center, AJA University of Medical Sciences, Tehran, Iran
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Saeed Oraee-Yazdani
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
158
|
Zhang YD, Yuan W, Bi H, Yang X, Zhang YY, Chen W. Whole-genome resequencing reveals collagen-related genes in Kele pigs. PLoS One 2024; 19:e0311417. [PMID: 39739914 DOI: 10.1371/journal.pone.0311417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 09/18/2024] [Indexed: 01/02/2025] Open
Abstract
OBJECTIVE To verify the accuracy of collagen-specific SNP mutation loci of Kele pigs selected by whole genome resequencing, and to excavate collagen-related genes of Kele pigs, so as to lay a foundation for further molecular selection. METHODS Based on whole genome resequencing, candidate genes related to collagen trait of Kele pig were screened for gene annotation. Through KEGG and GO enrichment analysis of differential genes, we selected four genes that may affect collagen trait of collagen pig, namely COL9A1, COL6A5, COL4A3 and COL4A4. Then 14 specific SNP sites were randomly selected from the four genes for sanger sequencing verification, and finally RT-qPCR was used to verify the expression levels of related genes in different tissues of Kele pigs. RESULTS Our sequencing results revealed that 241.04 G of clean data, Q30 reached 93.96% and the average coverage depth was 9.04×. After data analysis, the SNP annotation of Kele pigs identified 4,570 high-impact mutation sites that could result in protein function loss, with SNPs primarily distributed in the intronic and exonic regions. There were 132,256 middle-impact mutation sites and 318,150 low-impact mutation sites that could potentially impact protein properties. Additionally, The INDEL annotation results revealed a total of 17,806 high-impact mutation sites that could potentially result in the loss of protein function. There were 4740 medium-impact mutation sites that have the potential to affect protein properties, as well as 19,298 low-impact mutation sites. Furthermore, there were 14,197,763 mutation sites of modification influence degree in the analysis. In addition, through real-time fluorescence quantitative PCR results, we found that the expression levels of collagen-related genes COL9A1 and COL6A5 in skin tissues were higher than those in other tissues, and the expression levels of COL4A4 and COL4A3 in kidney tissues were higher than those in other tissues. The SNP site verification results showed that the 14 SNP mutation sites randomly selected by us were the same as the SNP mutation sites screened by whole genome resequencing. CONCLUSION A total of 307 genes related to collagen traits were excavated, including COL9A1, COL6A5, EP300, SOS2 and EPO, etc. It was found that COL9A1 and COL6A5 genes were significantly expressed in the skin tissue of Kele pigs, and COL4A4 and COL4A3 genes were significantly expressed in the kidney tissue of Kele pigs. The mutations of 14 randomly selected loci in the four related genes were consistent with the results of previous whole genome resequencing analysis, indicating that the specific SNP molecular marker information obtained by whole genome resequencing can be used as the basis for analyzing collagen traits of Kele pig. Our results are conducive to further research on collagen trait regulation of Kele pigs and development and utilization of Kele pigs in the future.
Collapse
Affiliation(s)
- Yu Dan Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, Guizhou Province, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Guiyang, Guizhou Province, China
- College of Animal Science, Guizhou University, Guiyang, Guizhou Province, China
| | - Wei Yuan
- College of Animal Science, Guizhou University, Guiyang, Guizhou Province, China
| | - Huan Bi
- Guizhou Agricultural Vocational College, Guiyang, Guizhou, China
| | - Xiao Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, Guizhou Province, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Guiyang, Guizhou Province, China
- College of Animal Science, Guizhou University, Guiyang, Guizhou Province, China
| | - Yi Yu Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, Guizhou Province, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Guiyang, Guizhou Province, China
- College of Animal Science, Guizhou University, Guiyang, Guizhou Province, China
| | - Wei Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, Guizhou Province, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Guiyang, Guizhou Province, China
- College of Animal Science, Guizhou University, Guiyang, Guizhou Province, China
| |
Collapse
|
159
|
Zheng Q, Lei FP, Hui S, Tong M, Liang LH. Ginsenoside Rb1 Relieves Cellular Senescence and Pulmonary Fibrosis by Promoting NRF2/QKI/SMAD7 Axis. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:2491-2509. [PMID: 39756830 DOI: 10.1142/s0192415x24500952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Cellular senescence is an adverse factor in the development of pulmonary fibrosis (PF). Ginsenoside Rb1 has been found to inhibit both cellular senescence and PF. This study aimed to elucidate the molecular mechanisms by which ginsenoside Rb1 regulates cellular senescence and PF. A PF mouse model was established by Bleomycin (BLM) administration, and a cell model of senescence was constructed using MRC-5 cells treated with Adriamycin RD (ARD) administration. Hematoxylin and Eosin (HE) staining and Masson staining were employed to evaluate cellular structure and collagen fiber content. RT-qPCR and western blotting were used to detect mRNA and protein expression of the target genes. Enzyme-linked Immunosorbent Assay (ELISA) was applied to measure the protein concentration of IL-1[Formula: see text] and IL-18. SA-[Formula: see text]-gal staining was used to evaluate cellular senescence. Our results show that ginsenoside Rb1 effectively suppressed BLM-induced PF in mice. ARD administration to induce cellular senescence reduced NRF2, QKI, and SMAD7 expression in MRC-5 cells. By inducing NRF2 overexpression, ARD-induced cellular senescence and fibrosis in MRC-5 cells were relieved. Notably, NRF2 knockdown abolished the mitigating effects of ginsenoside Rb1 on ARD-induced cellular senescence and fibrosis in MRC-5 cells. Mechanistically, NRF2 increased SMAD7 mRNA stability through the transcriptional regulation of QKI. As expected, ginsenoside Rb1 alleviated ARD-induced senescence and fibrosis in MRC-5 cells by activating the NRF2/QKI/SMAD7 axis. Therefore, it was found that ginsenoside Rb1 mitigates cellular senescence and fibrosis during PF progression by activating the NRF2/QKI/SMAD7 axis. This study provides a potential therapeutic strategy for the treatment of PF and elucidates its mechanism of action.
Collapse
Affiliation(s)
- Qing Zheng
- Department of Geriatrics, Hunan Provincial People's Hospital, (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, P. R. China
| | - Feng-Ping Lei
- Department of Geriatrics, Hunan Provincial People's Hospital, (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, P. R. China
| | - Shan Hui
- Department of Geriatrics, Hunan Provincial People's Hospital, (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, P. R. China
| | - Ming Tong
- Department of Infectious Diseases, Hunan Provincial People's Hospital, (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, P. R. China
| | - Li-Hui Liang
- Department of Geriatrics, Hunan Provincial People's Hospital, (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, P. R. China
| |
Collapse
|
160
|
Tan YY, Zhang DW, Yang C, Huang Y, Kang JY, Xu ZH, Wei YY, Ding ZX, Fei GH. ASIC1a regulates airway epithelial cell pyroptosis in acute lung injury by NLRP3-Caspase1-GSDMD pathway. Int Immunopharmacol 2024; 143:113623. [PMID: 39549550 DOI: 10.1016/j.intimp.2024.113623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/30/2024] [Accepted: 11/10/2024] [Indexed: 11/18/2024]
Abstract
BACKGROUND Acidosis is the most common complication that seriously affects the prognosis of acute respiratory distress syndrome (ARDS). Acid-sensitive ion channel 1a (ASIC1a) is activated in acidic environments to regulate inflammatory process. However, the role of ASIC1a in ARDS is unclear. METHODS In this study, we examined the expression of ASIC1a in airway epithelial cells in an acidic environment. We then investigated whether blocking ASIC1a could inhibit pyroptosis of airway epithelial cells and the molecular mechanism. In the mouse acute lung injury (ALI) model, we observed the changes of lung histopathology, arterial blood gas and pyroptosis related indexes after ASIC1a inhibition. Bronchoalveolar lavage fluid (BALF) from patients with ARDS were collected to explore the expression level of ASIC1a in ARDS patients. RESULTS Inhibiting ASIC1a can reduce the airway epithelial cell pyroptosis induced by an extracellular acidic environment. ASIC1a can bind to PRKACA, and silencing ASIC1a and PRKACA can inhibit the occurrence of pyroptosis in airway epithelial cells. Compared with control group, arterial blood pH and PaO2 in ALI group were significantly reduced. The inflammation in the lungs is more intense, and the mRNA and protein of NLRP3, Caspase1 and GSDMD were increased, while ASIC1a specific blocker psalmotoxin-1 alleviated this phenomenon. The expression of ASIC1a in BALF of ARDS patients was significantly increased, especially in non-survival group. CONCLUSION Acidic micro-environment can induce the increased expression of ASIC1a, and inhibition of ASIC1a can alleviate the inflammation and airway epithelial cell pyroptosis in ARDS. ASIC1a may be a new target for the treatment of ARDS.
Collapse
Affiliation(s)
- Yuan-Yuan Tan
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China; Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei 230022, Anhui Province, China
| | - Da-Wei Zhang
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China; Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei 230022, Anhui Province, China
| | - Chun Yang
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei 230022, Anhui Province, China; Department of Emergency Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yan Huang
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Jia-Ying Kang
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China; Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei 230022, Anhui Province, China
| | - Zhong-Hua Xu
- Center for Scientific Research, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuan-Yuan Wei
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China; Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei 230022, Anhui Province, China
| | - Zhen-Xing Ding
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei 230022, Anhui Province, China; Department of Emergency Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| | - Guang-He Fei
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China; Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei 230022, Anhui Province, China.
| |
Collapse
|
161
|
Ou H, Lin J, Ji L, Ye L, Ling M, Liao X, Lin F, Wang Y, Luo B, Hu Z, Pan L. Ferritinophagy mediated by the AMPK/ULK1 pathway is involved in ferroptosis subsequent to ventilator-induced lung injury. Respir Res 2024; 25:440. [PMID: 39719634 DOI: 10.1186/s12931-024-03076-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 12/18/2024] [Indexed: 12/26/2024] Open
Abstract
Mechanical ventilation (MV) remains a cornerstone of critical care; however, its prolonged application can exacerbate lung injury, leading to ventilator-induced lung injury (VILI). Although previous studies have implicated ferroptosis in the pathogenesis of VILI, the underlying mechanisms remain unclear. This study investigated the roles of ferritinophagy in ferroptosis subsequent to VILI. Using C57BL/6J mice and MLE-12 cells, we established both in vivo and in vitro models of VILI and cyclic stretching (CS)-induced cellular injury. We assessed lung injury and the biomarkers of ferroptosis and ferritinophagy, after appropriate pretreatments. This study demonstrated that high tidal volumes (HTV) for 4 h enhanced the sensitivity to ferroptosis in both models, evidenced by increased intracellular iron levels, lipid peroxidation and cell death, which can be mitigated by ferrostatin-1 treatment. Notably, nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy contributed to ferroptosis in VILI. Inhibition of autophagy with 3-methyladenine or NCOA4 knockdown decreased intracellular Fe2+ levels and inhibited lipid peroxidation, thereby attenuating CS-induced lung injury. Furthermore, it has also been observed that the AMPK/ULK1 axis can trigger ferritinophagy in VILI. Collectively, our study indicated that MV can induce ferroptosis by promoting NCOA4-dependent ferritinophagy, which could be a novel therapeutic target for the prevention and treatment of VILI.
Collapse
Affiliation(s)
- Huajin Ou
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, He Di Rd No.71, Nanning, 530021, P. R. China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
| | - Jinyuan Lin
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, He Di Rd No.71, Nanning, 530021, P. R. China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
| | - Liu Ji
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, He Di Rd No.71, Nanning, 530021, P. R. China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
| | - Liu Ye
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, He Di Rd No.71, Nanning, 530021, P. R. China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
| | - Maoyao Ling
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, He Di Rd No.71, Nanning, 530021, P. R. China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
| | - Xiaoting Liao
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, He Di Rd No.71, Nanning, 530021, P. R. China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
| | - Fei Lin
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, He Di Rd No.71, Nanning, 530021, P. R. China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
| | - Yuqing Wang
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, He Di Rd No.71, Nanning, 530021, P. R. China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
| | - Bijun Luo
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
- Department of Anesthesiology, The Maternal and Child Health Care Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Zhaokun Hu
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, He Di Rd No.71, Nanning, 530021, P. R. China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
| | - Linghui Pan
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, He Di Rd No.71, Nanning, 530021, P. R. China.
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China.
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China.
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China.
| |
Collapse
|
162
|
Zhou W, Wang Y, Ye P, Hu S, Li S, Wang M, Sheng D, Chen Y, Shen W, Zhang Y, Liu F, Zhang W, Lv X, Wang X, Yang H. Effects of Transcutaneous Electrical Acupoint Stimulation on the Incidence of Hypoxia in Elderly Patients Undergoing Painless Gastrointestinal Endoscopy: A Randomized Controlled Trial. Pain Res Manag 2024; 2024:1251246. [PMID: 39734602 PMCID: PMC11682864 DOI: 10.1155/prm/1251246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 10/20/2024] [Accepted: 11/27/2024] [Indexed: 12/31/2024]
Abstract
Background: Hypoxia is not uncommon in elderly patients during painless gastrointestinal endoscopy. This study aimed to determine the effectiveness of transcutaneous electrical acupoint stimulation (TEAS) in reducing the occurrence of hypoxia symptoms in elderly patients. Methods: Patients were randomly and equally grouped into sham control (n = 109) or TEAS group (n = 109) by using the random number table method. Patients in the TEAS group received electrical stimulation at the bilateral ST36 points 30 min before the examination until the end of the painless gastrointestinal endoscopy. Patients in the control group only had electrodes attached to bilateral nonacupoints in a similar pattern as the TEAS group without electrical stimulation. The primary endpoints measured were the incidence of hypoxia and severe hypoxia. The secondary endpoints included propofol dosage, sedation-related adverse events, hemodynamic parameters, surgical duration, patient recovery time, pain score, patient satisfaction, anesthesiologist satisfaction, and endoscopist satisfaction. Results: Of the 251 patients who participated in this study, 218 patients ended up completing the final study. The primary outcome was that, compared with group control, the incidence of hypoxia in group TEAS was reduced by 11% (19.3% vs. 8.3%, p=0.018) and the incidence of severe hypoxia did not show a significant change (7.3% vs. 2.8%, p=0.122). And there was a significant decrease in the occurrence of patients requiring emergency airway assistance (increased oxygen flow: 16.5% vs. 6.4%, p=0.019, jaw thrust: 11.0% vs. 3.7%, p=0.038, mask-assisted ventilation: 5.5% vs. 1.8%, p=0.015). Conclusion: TEAS can reduce the incidence of hypoxia in elderly patients undergoing painless gastrointestinal endoscopy. Trial Registration: ClinicalTrials.gov identifier: ChiCTR2200059465.
Collapse
Affiliation(s)
- Wenyu Zhou
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Department of Anesthesiology and Pain Management, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Yu Wang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Pengcheng Ye
- Department of Anesthesiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Song Hu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Siyu Li
- Department of Anesthesiology and Pain Management, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Mingxia Wang
- Department of Anesthesiology and Pain Management, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Duanyang Sheng
- Department of Anesthesiology and Pain Management, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Yuanli Chen
- Department of Anesthesiology and Pain Management, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Wang Shen
- Department of Anesthesiology and Pain Management, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Yi Zhang
- Department of Anesthesiology and Pain Management, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Feng Liu
- Department of Anesthesiology and Pain Management, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Wei Zhang
- Department of Anesthesiology and Pain Management, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Xin Lv
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Xiangrui Wang
- Department of Anesthesiology and Pain Management, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Hao Yang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| |
Collapse
|
163
|
Xia S, Gu X, Wang G, Zhong Y, Ma F, Liu Q, Xie J. Regulated Cell Death of Alveolar Macrophages in Acute Lung Inflammation: Current Knowledge and Perspectives. J Inflamm Res 2024; 17:11419-11436. [PMID: 39722732 PMCID: PMC11669335 DOI: 10.2147/jir.s497775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 11/29/2024] [Indexed: 12/28/2024] Open
Abstract
Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is a common and serious clinical lung disease characterized by extensive alveolar damage and inflammation leading to impaired gas exchange. Alveolar macrophages (AMs) maintain homeostatic properties and immune defenses in lung tissues. Several studies have reported that AMs are involved in and regulate ALI/ARDS onset and progression via different regulated cell death (RCD) programs, such as pyroptosis, apoptosis, autophagic cell death, and necroptosis. Notably, the effects of RCD in AMs in disease are complex and variable depending on the environment and stimuli. In this review, we provide a comprehensive perspective on how regulated AMs death impacts on ALI/ARDS and assess its potential in new therapeutic development. Additionally, we describe the crosstalk between different RCD types in ALI, and provide new perspectives for the treatment of ALI/ARDS and other severe lung diseases.
Collapse
Affiliation(s)
- Siwei Xia
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Xiaoyan Gu
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Gaojian Wang
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Yizhi Zhong
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Fengjie Ma
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Qinxue Liu
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Junran Xie
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| |
Collapse
|
164
|
Zhu M, Li N, Fan L, Wu R, Cao L, Ren Y, Lu C, Zhang L, Cai Y, Shi Y, Lin Z, Lu X, Leng J, Zhong S, Hu X, Huang B, Huang R, Zhou W, Yao D, Wu L, Wu W, Liu Q, Xia P, Chen R, Shi W, Zhang R, Lv S, Wang C, Yu L, Li J, Wang Q, Li K, Jin H. Single-cell transcriptomic and spatial analysis reveal the immunosuppressive microenvironment in relapsed/refractory angioimmunoblastic T-cell lymphoma. Blood Cancer J 2024; 14:218. [PMID: 39695118 DOI: 10.1038/s41408-024-01199-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
Angioimmunoblastic T-cell lymphoma (AITL) is a kind of aggressive T-cell lymphoma with significant enrichment of non-malignant tumor microenvironment (TME) cells. However, the complexity of TME in AITL progression is poorly understood. We performed single-cell RNA-Seq (scRNA-seq) and imaging mass cytometry (IMC) analysis to compare the cellular composition and spatial architecture between relapsed/refractory AITL (RR-AITL) and newly diagnosed AITL (ND-AITL). Our results showed that the malignant T follicular helper (Tfh) cells showed significantly increased proliferation driven by transcriptional activation of YY1 in RR-AITL, which is markedly associated with the poor prognosis of AITL patients. The CD8+ T cell proportion and cytotoxicity decreased in RR-AITL TME, resulting from elevated expression of the inhibitory checkpoints such as PD-1, TIGIT, and CTLA4. Notably, the transcriptional pattern of B cells in RR-AITL showed an intermediate state of malignant transformation to B-cell-lymphoma, and contributed to immune evasion by highly expressing CD47 and PD-L1. Besides, compared to ND-AITL samples, myeloid-cells-centered spatial communities were more prevalent but showed reduced phagocytic activity and impaired antigen processing and presentation in RR-AITL TME. Furthermore, specific inhibitory ligand-receptor interactions, such as CLEC2D-KLRB1, CTLA4-CD86, and MIF-CD74, were exclusively identified in the RR-AITL TME. Our study provides a high-resolution characterization of the immunosuppression ecosystem and reveals the potential therapeutic targets for RR-AITL patients.
Collapse
Affiliation(s)
- Mengyan Zhu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Lymphoma Center, Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ning Li
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Department of Hematology of the Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Northern Jiangsu Institute of Clinical Medicine, Huai'an, Jiangsu, China
| | - Lei Fan
- Lymphoma Center, Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China
| | - Rongrong Wu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Cao
- Lymphoma Center, Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China
| | - Yimin Ren
- Lymphoma Center, Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China
| | - Chuanyang Lu
- Department of Hematology of the Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Northern Jiangsu Institute of Clinical Medicine, Huai'an, Jiangsu, China
| | - Lishen Zhang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yun Cai
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuzhu Shi
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zihan Lin
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xueying Lu
- Lymphoma Center, Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China
| | - Jiayan Leng
- Lymphoma Center, Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Shiyang Zhong
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xingfei Hu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Bin Huang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Runheng Huang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wanting Zhou
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Diru Yao
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lingxiang Wu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wei Wu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Quanzhong Liu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Peng Xia
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ruize Chen
- Lymphoma Center, Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China
| | - Wenyu Shi
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China
| | - Ruohan Zhang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Sali Lv
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chunling Wang
- Department of Hematology of the Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Northern Jiangsu Institute of Clinical Medicine, Huai'an, Jiangsu, China
| | - Liang Yu
- Department of Hematology of the Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Northern Jiangsu Institute of Clinical Medicine, Huai'an, Jiangsu, China
| | - Jianyong Li
- Lymphoma Center, Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China.
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China.
| | - Qianghu Wang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China.
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China.
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China.
- Biomedical Big Data Center, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Kening Li
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China.
- Department of Hematology of the Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Northern Jiangsu Institute of Clinical Medicine, Huai'an, Jiangsu, China.
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China.
- Biomedical Big Data Center, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Hui Jin
- Lymphoma Center, Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China.
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
165
|
Li Y, He Y. Therapeutic applications of stem cell-derived exosomes in radiation-induced lung injury. Cancer Cell Int 2024; 24:403. [PMID: 39695650 DOI: 10.1186/s12935-024-03595-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
Radiation-induced lung injury is a common complication of chest tumor radiotherapy; however, effective clinical treatments are still lacking. Stem cell-derived exosomes, which contain various signaling molecules such as proteins, lipids, and miRNAs, not only retain the tissue repair and reconstruction properties of stem cells but also offer improved stability and safety. This presents significant potential for treating radiation-induced lung injury. Nonetheless, the clinical adoption of stem cell-derived exosomes for this purpose remains limited due to scientific, practical, and regulatory challenges. In this review, we highlight the current pathology and therapies for radiation-induced lung injury, focusing on the potential applications and therapeutic mechanisms of stem cell-derived exosomes. We also discuss the limitations of existing stem cell-derived exosomes and outline future directions for exosome-based treatments for radiation-induced lung injury.
Collapse
Affiliation(s)
- Ying Li
- Department of Radiotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yan He
- Department of Radiotherapy, West China Hospital, Sichuan University, Chengdu, China.
- Department of Cancer Center, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
166
|
Saleem M, Shahzad KA, Marryum M, Singh S, Zhou Q, Du S, Wang S, Shao C, Shaikh II. Exosome-based therapies for inflammatory disorders: a review of recent advances. Stem Cell Res Ther 2024; 15:477. [PMID: 39695750 DOI: 10.1186/s13287-024-04107-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024] Open
Abstract
Exosomes, small extracellular vesicles secreted by cells, have emerged as focal mediators in intercellular communication and therapeutic interventions across diverse biomedical fields. Inflammatory disorders, including inflammatory bowel disease, acute liver injury, lung injury, neuroinflammation, and myocardial infarction, are complex conditions that require innovative therapeutic approaches. This review summarizes recent advances in exosome-based therapies for inflammatory disorders, highlighting their potential as diagnostic biomarkers and therapeutic agents. Exosomes have shown promise in reducing inflammation, promoting tissue repair, and improving functional outcomes in preclinical models of inflammatory disorders. However, further research is needed to overcome the challenges associated with exosome isolation, characterization, and delivery, as well as to fully understand their mechanisms of action. Current limitations and future directions in exosome research underscore the need for enhanced isolation techniques and deeper mechanistic insights to harness exosomes' full therapeutic potential in clinical applications. Despite these challenges, exosome-based therapies hold great potential for the treatment of inflammatory disorders and may offer a new paradigm for personalized medication.
Collapse
Affiliation(s)
- Mavra Saleem
- Department of Zoology, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Khawar Ali Shahzad
- Department of Zoology, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
- Department of ORL-HNS, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Munazzah Marryum
- Department of Zoology, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Shekhar Singh
- Lishui People's Hospital, Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui, 323000, Zhejiang, China
| | - Quan Zhou
- Lishui People's Hospital, Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui, 323000, Zhejiang, China
| | - Siting Du
- Lishui People's Hospital, Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui, 323000, Zhejiang, China
| | - Shuanghu Wang
- Lishui People's Hospital, Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui, 323000, Zhejiang, China
| | - Chuxiao Shao
- Lishui People's Hospital, Central Laboratory of The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, Zhejiang, China
| | - Imran Ibrahim Shaikh
- Lishui People's Hospital, Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui, 323000, Zhejiang, China.
| |
Collapse
|
167
|
Tan R, Ge C, Yan Y, Guo H, Han X, Zhu Q, Du Q. Deciphering ferroptosis in critical care: mechanisms, consequences, and therapeutic opportunities. Front Immunol 2024; 15:1511015. [PMID: 39737174 PMCID: PMC11682965 DOI: 10.3389/fimmu.2024.1511015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 12/03/2024] [Indexed: 01/01/2025] Open
Abstract
Ischemia-reperfusion injuries (IRI) across various organs and tissues, along with sepsis, significantly contribute to the progression of critical illnesses. These conditions disrupt the balance of inflammatory mediators and signaling pathways, resulting in impaired physiological functions in human tissues and organs. Ferroptosis, a distinct form of programmed cell death, plays a pivotal role in regulating tissue damage and modulating inflammatory responses, thereby influencing the onset and progression of severe illnesses. Recent studies highlight that pharmacological agents targeting ferroptosis-related proteins can effectively mitigate oxidative stress caused by IRI in multiple organs, alleviating associated symptoms. This manuscript delves into the mechanisms and signaling pathways underlying ferroptosis, its role in critical illnesses, and its therapeutic potential in mitigating disease progression. We aim to offer a novel perspective for advancing clinical treatments for critical illnesses.
Collapse
Affiliation(s)
- Ruimin Tan
- School of Clinical Medical, North China University of Science and Technology, Tangshan, Hebei, China
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Chen Ge
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Yating Yan
- School of Clinical Medical, North China University of Science and Technology, Tangshan, Hebei, China
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - He Guo
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
- School of Graduate, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xumin Han
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
- School of Graduate, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Qiong Zhu
- Department of Orthopaedics, The People’s Hospital Of Shizhu, Chongqing, China
| | - Quansheng Du
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
| |
Collapse
|
168
|
Theodoropoulou MA, Mantzourani C, Kokotos G. Histone Deacetylase (HDAC) Inhibitors as a Novel Therapeutic Option Against Fibrotic and Inflammatory Diseases. Biomolecules 2024; 14:1605. [PMID: 39766311 PMCID: PMC11674560 DOI: 10.3390/biom14121605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/08/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Histone deacetylases (HDACs) are enzymes that play an essential role in the onset and progression of cancer. As a consequence, a variety of HDAC inhibitors (HDACis) have been developed as potent anticancer agents, several of which have been approved by the FDA for cancer treatment. However, recent accumulated research results have suggested that HDACs are also involved in several other pathophysiological conditions, such as fibrotic, inflammatory, neurodegenerative, and autoimmune diseases. Very recently, the HDAC inhibitor givinostat has been approved by the FDA for an indication beyond cancer: the treatment of Duchenne muscular dystrophy. In recent years, more and more HDACis have been developed as tools to understand the role that HDACs play in various disorders and as a novel therapeutic approach to fight various diseases other than cancer. In the present perspective article, we discuss the development and study of HDACis as anti-fibrotic and anti-inflammatory agents, covering the period from 2020-2024. We envision that the discovery of selective inhibitors targeting specific HDAC isozymes will allow the elucidation of the role of HDACs in various pathological processes and will lead to the development of promising treatments for such diseases.
Collapse
Affiliation(s)
- Maria A. Theodoropoulou
- Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece; (M.A.T.); (C.M.)
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Christiana Mantzourani
- Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece; (M.A.T.); (C.M.)
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - George Kokotos
- Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece; (M.A.T.); (C.M.)
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, 15771 Athens, Greece
| |
Collapse
|
169
|
Qi D, Zhang H, Xiong F, Zhang G, Tao B, Wang C. Renal cell carcinoma and macrophage research: A bibliometric analysis (2004-2023). Medicine (Baltimore) 2024; 103:e40954. [PMID: 39686418 PMCID: PMC11651438 DOI: 10.1097/md.0000000000040954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
To analyze hotspots and trends in renal cell carcinoma (RCC)-macrophage research using bibliometric analysis, although numerous studies on macrophages in RCC have been recently reported, understanding the progressive trends in this field remains challenging. Publications focused on macrophages in RCC were extracted from the Web of Science Core Collection. VOSviewer, Citespace, and Bibliometrics online platforms were used to visualize hot topics and global trends in RCC-macrophage research. In total, 778 papers were collected. China produced the most articles; however, the United States accounted for the largest number of citations. Oncology journals published the most articles, and these were cited most frequently. Based on keyword analysis, "prognosis," "immunotherapy," "tumor microenvironment," and "immune infiltration" represented the primary research hotspots. In summary, RCC-macrophage studies have emerged as a key research focus; particularly, incorporating multiomics data and applying artificial intelligence for predictive modeling have demonstrated significant potential. Our study suggests that the resistance mechanism of immune checkpoint inhibitors and the interaction between macrophages and immune checkpoint inhibitors will be pivotal areas for future research.
Collapse
Affiliation(s)
- Dingtian Qi
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Haoxun Zhang
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Feng Xiong
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guoling Zhang
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Boju Tao
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chunyang Wang
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
170
|
Al-Sherief S, El-Hadidy A, Hamed S, El-Hawwary A, Mazroa S. Bone marrow mesenchymal stem cells (BM-MSCs) modulate MMP9 expression and promote articular cartilage regeneration in knee joint of a model of arthritis induced in adult rat: histological and immunohistochemical study. J Mol Histol 2024; 56:38. [PMID: 39661261 DOI: 10.1007/s10735-024-10284-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 10/31/2024] [Indexed: 12/12/2024]
Abstract
Arthritis is characterized by the progressive degeneration of articular cartilage, and the avascular nature of cartilage limits its capacity for self-repair. Stem cells are considered a promising treatment option due to their multipotent differentiation potential. The aim of this work was to investigate the structural changes in the hyaline articular cartilage of the knee joint in a model of arthritis induced by complete Freund's adjuvant, and to assess intra-articular injection of bone marrow mesenchymal stem cells (BM-MSCs) through both histological and immunohistochemical study. Adult male albino rats were divided into four groups: group 0 (donor group), group I (control group), group II (arthritis group) and group III (BM-MSCs treated arthritis group). Samples were collected 2, 6 and 10 weeks after the onset of the experiment. Sections were stained with; hematoxylin and eosin, Safranin O fast green stain, Masson's trichrome stain and anti-MMP9 antibody. In Group II (arthritis group), the articular cartilage showed signs of degeneration, including chondrocyte extensive proliferation, fibrillations, fissuring, and denudation, with fibrous tissue covering the exposed surface. There was a significant decrease in cartilage thickness, collagen content, and proteoglycan levels. The integrated density of MMP9 in the cartilage was significantly increased compared to Group I (control group). In contrast, Group III (BM-MSCs-treated arthritis group) exhibited a continuous cartilage surface with no cracks or fissures. There was a significant increase in cartilage thickness, collagen content, and proteoglycan levels, while the integrated density of MMP9 was significantly decreased compared to Group II (arthritis group).
Collapse
Affiliation(s)
- Sara Al-Sherief
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | - Azza El-Hadidy
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Shereen Hamed
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Amany El-Hawwary
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Shireen Mazroa
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
171
|
Coppolino F, Brunetti S, Bottazzo LM, Cosenza G, Sansone P, Fiore M, Passavanti MB, Pota V, Pace MC. Adding Dexmedetomidine to Methylene Blue in Thoracic Paravertebral Block for Video-Assisted Lobectomy: A Case Series Study. Local Reg Anesth 2024; 17:99-105. [PMID: 39677514 PMCID: PMC11646378 DOI: 10.2147/lra.s487981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 11/08/2024] [Indexed: 12/17/2024] Open
Abstract
PURPOSE Thoracic surgery often results in severe chronic postoperative pain. Current evidence favors two locoregional techniques. Thoracic Epidural Anesthesia (TEA), the gold standard, and Thoracic Paravertebral Block (TPVB), which is associated with fewer side effects but is limited by short duration of action of local anesthetics (LA) and potential failure due to improper drug distribution. This study investigates the use of dexmedetomidine (DEX) as adjuvant to prolong the effects of LA in TPVB, with methylene blue used for visual confirmation of accurate injectate spread. PATIENTS AND METHODS We observed 6 patients undergoing Video-Assisted Thoracoscopy (VATS) lobectomy who received TPVB with ropivacaine, DEX and methylene blue. The primary endpoint was postoperative pain recorded at 1, 12, 24, 48 hours using Numeric Rating Scale (NRS); the secondary endpoints were cumulative opioid consumption in the first 24 hours in Milligram Morphine Equivalents (MME); adverse events: occurrence of bradycardia, hypotension, Postoperative Nausea and Vomiting (PONV); length of hospital stay. All patients completed the study. RESULTS Our results showed optimal pain scores, with NRS scores always below 4, decreased need for opioids, and prolonged analgesia. None of the patients had bradycardia nor PONV, but two patients experienced acute and self-limited hypotension following TPVB. CONCLUSION Thoracic Paravertebral Block with Dexmedetomidine and methylene blue was effective and safe in controlling postoperative pain. Methylene blue could help improving knowledge on anesthetics distribution to reduce failure rates.
Collapse
Affiliation(s)
- Francesco Coppolino
- Department of Women, Children and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Simona Brunetti
- Department of Women, Children and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Leonardo Maria Bottazzo
- Department of Women, Children and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Gianluigi Cosenza
- Department of Women, Children and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Pasquale Sansone
- Department of Women, Children and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Marco Fiore
- Department of Women, Children and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Maria Beatrice Passavanti
- Department of Women, Children and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Vincenzo Pota
- Department of Women, Children and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Maria Caterina Pace
- Department of Women, Children and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
172
|
Yang F, Wang J, Zhang H, Zhang Y, Yang W, Gao R, Yu J, Chen X, Ma H. Effect of Intravenous Lidocaine Infusion on Propofol Dose and Perioperative Pain During Moderate Sedation-Analgesia for Hysteroscopy: A Randomized Controlled Trial. Drug Des Devel Ther 2024; 18:5873-5880. [PMID: 39664966 PMCID: PMC11633289 DOI: 10.2147/dddt.s484486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 11/30/2024] [Indexed: 12/13/2024] Open
Abstract
Purpose In China, the majority of hysteroscopic procedures require moderate sedation and analgesia. The efficacy of intravenous lidocaine in reducing the need for sedatives and alleviating perioperative pain during hysteroscopy remains equivocal. This study aims to determine whether the intravenous administration of lidocaine can reduce the required dose of propofol and enhance perioperative pain management. Patients and Methods We conducted a prospective, single-center, double-blind randomized controlled trial involving patients with ASA I-II undergoing hysteroscopy. Forty patients were randomly assigned in a 1:1 ratio to either receive an intravenous bolus dose of 1.5 mg/kg lidocaine, followed by a continuous intravenous infusion at 4 mg/kg/h until the conclusion of the procedure, or an equivalent volume of normal saline. Propofol was then titrated to maintain a MOAA/S score of ≤ 2. Results Compared with the control group, the lidocaine group showed a 13.8% decrease in the total dose of propofol (140.0[120.0, 155.0] mg vs 162.5[140.0, 197.5] mg), which was statistically significant (P = 0.014). The induction dose of propofol was 1.37 (1.29, 1.56) mg/kg in the lidocaine group and 1.61 (1.48, 1.94) mg/kg in the control group, respectively (P = 0.001). However, no significant differences were observed between the groups regarding the supplemental dose of propofol (P = 0.062), the number of involuntary movements during hysteroscopy (P = 0.384), or postoperative pain scores (T0: P = 0.628; T1: P = 0.886; T2: P = 0.711). Additionally, the incidence of intraoperative hypoxia (P = 1.000) and fatigue scores (T0: P = 0.878; T1: P = 0.401; T2: P = 0.056) between the two groups were not statistically significant. Conclusion Intravenous lidocaine reduces the dose requirements of propofol during the induction phase of anesthesia. However, it does not have a significant influence on alleviating intraoperative and postoperative pain during hysteroscopic procedures.
Collapse
Affiliation(s)
- Fan Yang
- Department of Anesthesiology, General Hospital of Ningxia Medical University, Yinchuan, 75004, People’s Republic of China
| | - Jie Wang
- Department of Anesthesiology, General Hospital of Ningxia Medical University, Yinchuan, 75004, People’s Republic of China
- Department of Anesthesiology, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, 650051, People’s Republic of China
| | - Huiwen Zhang
- Department of Anesthesiology, General Hospital of Ningxia Medical University, Yinchuan, 75004, People’s Republic of China
| | - Yonghai Zhang
- Department of Anesthesiology, General Hospital of Ningxia Medical University, Yinchuan, 75004, People’s Republic of China
| | - Wanji Yang
- Department of Anesthesiology, General Hospital of Ningxia Medical University, Yinchuan, 75004, People’s Republic of China
| | - Ran Gao
- Department of Anesthesiology, General Hospital of Ningxia Medical University, Yinchuan, 75004, People’s Republic of China
| | - Jingfang Yu
- Department of Anesthesiology, General Hospital of Ningxia Medical University, Yinchuan, 75004, People’s Republic of China
| | - Xuexin Chen
- Department of Anesthesiology, General Hospital of Ningxia Medical University, Yinchuan, 75004, People’s Republic of China
| | - Hanxiang Ma
- Department of Anesthesiology, General Hospital of Ningxia Medical University, Yinchuan, 75004, People’s Republic of China
| |
Collapse
|
173
|
Rao X, Luo H, Luo K, Hu C. Silencing SMAD4 inhibits inflammation and ferroptosis in asthma by blocking the IL-17A signaling pathway. Respir Res 2024; 25:429. [PMID: 39643876 PMCID: PMC11622552 DOI: 10.1186/s12931-024-03052-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 11/23/2024] [Indexed: 12/09/2024] Open
Abstract
BACKGROUND The TGF-β/SMAD signaling pathway is crucial in the pathogenesis of asthma. However, SMAD family member 4 (SMAD4), a key mediator of TGF-β, its roles and underlying mechanisms in asthma remain unclear. METHODS The in vivo and in vitro roles of SMAD4 in asthma were investigated through an ovalbumin (OVA)-induced mouse model and an interleukin-13 (IL-13)-induced cell model. The molecular mechanism of SMAD4 influenced asthma was examined using transcriptome sequencing, followed by feedback experiments involving recombinant human interleukin 17 A (rhIL-17 A), an IL-17 A signaling pathway activator. RESULTS SMAD4 was highly expressed in the asthma models. SMAD4 silencing alleviated damage to lung tissue and decreased inflammatory infiltration. Expression levels of Caspase-3, IgG, and inflammatory factors were reduced after silencing SMAD4. Silencing SMAD4 suppressed ferroptosis. Silencing SMAD4 also enhanced IL-13-induced BEAS-2B cell proliferation and suppressed apoptosis. Furthermore. IL-17 A signaling pathway was promoted in the asthma models, as evidenced by elevated IL-17RA, IL-17 A, and Act1 protein levels. SMAD4 silencing inhibited the expression levels of these IL-17 A pathway-associated proteins. Moreover, rhIL-17 A treatment notably reversed the impacts of SMAD4 silencing on asthma in the IL-13-induced cell model and OVA-induced mouse model, indicating that silencing SMAD4 inhibited inflammation and ferroptosis in asthma by blocking the IL-17 A signaling pathway. CONCLUSION Silencing SMAD4 prevents inflammation and ferroptosis in asthma by inhibiting the IL-17 pathway, which provides a novel potential approach for asthma therapy.
Collapse
Affiliation(s)
- Xingyu Rao
- Department of Pediatrics, First Affiliated Hospital of Gannan Medical University, No.128, Jinling Road, Zhanggong District, Ganzhou, 341000, China
| | - Hong Luo
- Department of Pediatrics, First Affiliated Hospital of Gannan Medical University, No.128, Jinling Road, Zhanggong District, Ganzhou, 341000, China
| | - Kaiyuan Luo
- Department of Pediatrics, First Affiliated Hospital of Gannan Medical University, No.128, Jinling Road, Zhanggong District, Ganzhou, 341000, China
| | - Chaohua Hu
- Department of Surgery I, The Third Affiliated Hospital of Gannan Medical University/Affiliated Stomatological Hospital, No. 46, Jingjiu Road, Zhanggong District, Ganzhou, Jiangxi Province, 341000, China.
| |
Collapse
|
174
|
Cheng S, Li Y, Sun X, Liu Z, Guo L, Wu J, Yang X, Wei S, Wu G, Xu S, Yang F, Wu J. The impact of glucose metabolism on inflammatory processes in sepsis-induced acute lung injury. Front Immunol 2024; 15:1508985. [PMID: 39712019 PMCID: PMC11659153 DOI: 10.3389/fimmu.2024.1508985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 11/18/2024] [Indexed: 12/24/2024] Open
Abstract
Acute lung injury (ALI) is a prevalent and critical complication of sepsis, marked by high incidence and mortality rates, with its pathogenesis still not being fully elucidated. Recent research has revealed a significant correlation between the metabolic reprogramming of glucose and sepsis-associated ALI (S-ALI). Throughout the course of S-ALI, immune cells, including macrophages and dendritic cells, undergo metabolic shifts to accommodate the intricate demands of immune function that emerge as sepsis advances. Indeed, glucose metabolic reprogramming in S-ALI serves as a double-edged sword, fueling inflammatory immune responses in the initial stages and subsequently initiating anti-inflammatory responses as the disease evolves. In this review, we delineate the current research progress concerning the pathogenic mechanisms linked to glucose metabolic reprogramming in S-ALI, with a focus on the pertinent immune cells implicated. We encapsulate the impact of glucose metabolic reprogramming on the onset, progression, and prognosis of S-ALI. Ultimately, by examining key regulatory factors within metabolic intermediates and enzymes, We have identified potential therapeutic targets linked to metabolic reprogramming, striving to tackle the inherent challenges in diagnosing and treating Severe Acute Lung Injury (S-ALI) with greater efficacy.
Collapse
Affiliation(s)
- Shilei Cheng
- School of Anesthesiology, Shandong Second Medical University, Weifang, China
| | - Yufei Li
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Anesthesiology, Jinan, China
- School of Pharmacy, Shandong University of Traditional Chinese Medicine (TCM), Jinan, China
| | - Xiaoliang Sun
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhirui Liu
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Liang Guo
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Anesthesiology, Jinan, China
| | - Jueheng Wu
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Xiaohan Yang
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Sisi Wei
- Department of Anesthesiology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, China
| | - Guanghan Wu
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Anesthesiology, Jinan, China
| | - Shilong Xu
- School of Anesthesiology, Shandong Second Medical University, Weifang, China
| | - Fan Yang
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Anesthesiology, Jinan, China
| | - Jianbo Wu
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Anesthesiology, Jinan, China
| |
Collapse
|
175
|
Liu SH, Zhang J, Zuo YG. Macrophages in inflammatory skin diseases and skin tumors. Front Immunol 2024; 15:1430825. [PMID: 39703508 PMCID: PMC11656021 DOI: 10.3389/fimmu.2024.1430825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 11/18/2024] [Indexed: 12/21/2024] Open
Abstract
Macrophages, as specialized, long-lasting phagocytic cells of the innate immune system, have garnered increasing attention due to their wide distribution and various functions. The skin, being the largest immune organ in the human body, presents an intriguing landscape for macrophage research, particularly regarding their roles in inflammatory skin diseases and skin tumors. In this review, we compile the latest research on macrophages in conditions such as atopic dermatitis, psoriasis, systemic sclerosis, systemic lupus erythematosus, rosacea, bullous pemphigoid, melanoma and cutaneous T-cell lymphoma. We aim to contribute to illustrating the pathogenesis and potential new therapies for inflammatory skin diseases and skin tumors from the perspective of macrophages.
Collapse
Affiliation(s)
| | | | - Ya-Gang Zuo
- Department of Dermatology, State Key Laboratory of Complex Severe and Rare Diseases, National Clinical Research Center for Dermatologic and Immunologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
176
|
Luo L, Liu K, Deng L, Wang W, Lai T, Li X. Chicoric acid acts as an ALOX15 inhibitor to prevent ferroptosis in asthma. Int Immunopharmacol 2024; 142:113187. [PMID: 39298822 DOI: 10.1016/j.intimp.2024.113187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/27/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Chicoric acid (CA) is a crucial immunologically active compound found in chicory and echinacea, possessing a range of biological activities. Ferroptosis, a type of iron-dependent cell death induced by lipid peroxidation, plays a key role in the development and advancement of asthma. Targeting ferroptosis could be a potential therapeutic strategy for treating asthma. PURPOSE The purpose of this study was to explore the screening of ALOX15, a pivotal target of ferroptosis in asthma, and potential therapeutic agents, as well as to investigate the promising potential of CA as an ALOX15 inhibitor for modulating ferroptosis in asthma. METHODS Through high-throughput data processing of bronchial epithelial RNA from asthma patients using bioinformatics and machine learning, the key target of ferroptosis in asthma, ALOX15, was identified. An inhibitor of ALOX15 was then obtained through high-throughput molecular docking and molecular dynamics simulation tests. In vitro experiments were conducted using a 16HBE cell model induced by house dust mite (HDM) and lipopolysaccharide (LPS), which were treated with the ALOX15 inhibitor (PD146176), CA treatment, or ALOX15 knockdown. In vivo experiments were also carried out using a mouse model induced by HDM and LPS. RESULTS The composite model of ALOX15 and CA in molecular dynamics simulations shows good stability and flexibility. Network pharmacological analysis reveals that CA regulates ferroptosis through ALOX15 in treating asthma. In vitro studies show that ALOX15 is highly expressed in HDM and LPS treatments, while CA inhibits HDM and LPS-induced ferroptosis in 16HBE cells by reducing ALOX15 expression. Knockdown of ALOX15 has the opposite effect. Metabolomics analysis identifies key compounds associated with ferroptosis, including L-Targinine, eicosapentaenoic acid, 16-hydroxy hexadecanoic acid, and succinic acid. In vivo experiments demonstrate that CA suppresses ALOX15 expression, inhibits ferroptosis, and improves asthma symptoms in mice. CONCLUSION Our research initially identified CA as a promising asthma treatment that effectively blocks ferroptosis by specifically targeting ALOX15. This study not only highlights CA as a potential therapeutic agent for asthma but also introduces novel targets and treatment options for this condition, along with innovative approaches for utilizing natural compounds to target diseases associated with ferroptosis.
Collapse
Affiliation(s)
- Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine. Guangdong Medical University, Zhanjiang, Guangdong 524023, China.
| | - Kangdi Liu
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine. Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Liyan Deng
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine. Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Wenjian Wang
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine. Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Tianli Lai
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine. Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Xiaoling Li
- Experimental Animal Center, Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| |
Collapse
|
177
|
Wada Y, Naito T, Fukushima T, Saito M. Evaluation of ALKBH2 and ALKBH3 gene regulation in patients with adult T-cell leukemia/lymphoma. Virol J 2024; 21:316. [PMID: 39633427 PMCID: PMC11619432 DOI: 10.1186/s12985-024-02590-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Human T-cell leukemia virus type 1 (HTLV-1) is an oncogenic virus that causes malignant adult T-cell leukemia/lymphoma (ATL). Patients infected with HTLV-1 are considered HTLV-1 carriers, and a small proportion of patients progress to life-threatening ATL after a long asymptomatic phase. No antiviral agent or preventive vaccine specific for HTLV-1 infection is established in current situation. For development of countermeasures to combat HTLV-1 infection and ATL, it is essential to expand our knowledge about their pathogenesis. Recently, AlkB homolog (ALKBH) family have been shown to participate in the oncogenesis of various cancer types. METHODS To investigate the potential role of ALKBH family members in the pathogenesis of ATL, we analyzed their gene expression dynamics in HTLV-1-infected T-cell lines and peripheral blood mononuclear cell-derived clinical specimens obtained from asymptomatic HTLV-1 carriers and patients with acute-type ATL. Epigenetic analysis was performed to dissect the mechanisms of ALKBH3 gene regulation using cultivated cells and a public dataset. RESULTS The mRNA expression levels of ALKBH2 and ALKBH3 were significantly or suggestively decreased in asymptomatic HTLV-1 carriers, but reverted in acute-type ATL patients, correlating with HTLV-1 basic leucine zipper factor gene expression. Intriguingly, the pre-mRNA expression of ALKBH2 and ALKBH3 was significantly suppressed in patients infected with HTLV-1, but not in healthy controls. Epigenetic analysis was performed to dissect the mechanisms of ALKBH3 gene regulation. In vitro analysis suggested a possible relationship between DNA methylation and ALKBH3 gene expression. Investigation of a public dataset revealed that specific CpG sites exhibited characteristically regulated methylation states in HTLV-1-infected T-cell subsets. CONCLUSION We discovered dynamically regulated patterns of ALKBH2 and ALKBH3 gene expression in patients infected with HTLV-1, and specific CpG sites epigenetically regulated by HTLV-1 infection. This study provides novel insights into HTLV-1 infection and contributes to the elucidation of ATL pathogenesis.
Collapse
Affiliation(s)
- Yuji Wada
- Department of Microbiology, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Tadasuke Naito
- Department of Microbiology, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Takuya Fukushima
- Laboratory of Hematoimmunology, Graduate School of Health Sciences, Faculty of Medicine, University of the Ryukyus, Nishihara, Okinawa, Japan
| | - Mineki Saito
- Department of Microbiology, Kawasaki Medical School, Kurashiki, Okayama, Japan.
| |
Collapse
|
178
|
Kostyuk SV, Malinovskaya EM, Umriukhin PE, Proskurnina EV, Ershova ES, Kameneva LV, Savinova EA, Kostyuk SE, Voronov II, Kraevaya OA, Troshin PA, Salimova TA, Kutsev SI, Veiko NN. Cytoprotective Effects and Intranuclear Localization of Sulfur-Containing Derivative of Buckminsterfullerene. FRONT BIOSCI-LANDMRK 2024; 29:408. [PMID: 39735985 DOI: 10.31083/j.fbl2912408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND There is a growing interest in exploring the biological characteristics of nanoparticles and exploring their potential applications. However, there is still a lack of research into the potential genotoxicity of fullerene derivatives and their impact on gene expression in human cells. In this study, we investigated the effects of a water-soluble fullerene derivative, C60[C6H4SCH2COOK]5H (F1), on human embryonic lung fibroblasts (HELF). METHODS 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) test was used to study the cytotoxicity of F1; reactive oxygen species (ROS) level was determined with 2,7-DCFH-DA; gene expression level was evaluated by reverse transcription polymerase chain reaction (RT-PCR); protein expression level was determined by flow cytofluorometry; fluorescence microscopy was used for visualization; Mann-Whitney statistical U-test was used for data processing. The differences were considered significant at p < 0.01. RESULTS F1 at a concentration of 0.3 mg/mL causes a short-term (up to 1 hour) increase in the number of double-strand breaks and oxidative DNA damage in HELF. Within 1 to 24 hours, F1 penetrates through the cell and nuclear membrane of HELF and localizes in the nucleus. In this case, the response of cells to DNA damage is activated: the functional activity of DNA repair genes, antioxidant and anti-apoptotic genes is increased within 24 hours. Due to the processes of activation of cell division and inhibition of apoptosis, an increase in the population of HELF cells in the presence of the fullerene derivative F1 is observed. F1 has a stabilizing effect on cell nuclei under the action of 1 Gy radiation. CONCLUSIONS An increase in antioxidant protection, activation of repair genes, anti-apoptotic genes, progression of the cell cycle, and a decrease in the level of oxidative damage, and DNA breaks in cells indicates the cytoprotective properties of F1.
Collapse
Affiliation(s)
| | | | - Pavel E Umriukhin
- Research Centre for Medical Genetics, 115522 Moscow, Russia
- M. Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| | | | | | | | | | | | - Ilya I Voronov
- Department of Kinetics and Catalysis, Federal Research Center for Problems of Chemical Physics and Medicinal Chemistry of RAS, 142432 Chernogolovka, Russia
| | - Olga A Kraevaya
- Department of Kinetics and Catalysis, Federal Research Center for Problems of Chemical Physics and Medicinal Chemistry of RAS, 142432 Chernogolovka, Russia
| | - Pavel A Troshin
- Department of Kinetics and Catalysis, Federal Research Center for Problems of Chemical Physics and Medicinal Chemistry of RAS, 142432 Chernogolovka, Russia
- Zhengzhou Research Institute of Harbin Institute of Technology, 450000 Zhengzhou, Henan, China
| | | | | | | |
Collapse
|
179
|
He S, Jiang X, Yang J, Wu Y, Shi J, Wu X, Du S, Zhang Y, Gong L, Dong S, Yu J. Nicotinamide mononucleotide alleviates endotoxin-induced acute lung injury by modulating macrophage polarization via the SIRT1/NF-κB pathway. PHARMACEUTICAL BIOLOGY 2024; 62:22-32. [PMID: 38100537 PMCID: PMC10732210 DOI: 10.1080/13880209.2023.2292256] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 12/03/2023] [Indexed: 12/17/2023]
Abstract
CONTEXT Sepsis-induced acute lung injury (ALI) is a severe condition with limited effective therapeutics; nicotinamide mononucleotide (NMN) has been reported to exert anti-inflammatory activities. OBJECTIVE This study explores the potential mechanisms by which NMN ameliorates sepsis-induced ALI in vivo and in vitro. MATERIALS AND METHODS Cultured MH-S cells and a murine model were used to evaluate the effect of NMN on sepsis-induced ALI. MH-S cells were stimulated with LPS (1 μg/mL) and NMN (500 μM) for 12 h grouping as control, LPS, and LPS + NMN. Cell viability, apoptotic status, and M1/2 macrophage-related markers were detected. The mice were pretreated intraperitoneally with NMN (500 mg/kg) and/or EX-527 (5 mg/kg) 1 h before LPS injection and randomized into 7 groups (n = 8): control, LPS, LPS + NMN, NMN, LPS + NMN + EX-527 (a SIRT1 inhibitor), LPS + EX-527, and EX-527. After 12 h, lung histopathology, W/D ratio, MPO activity, NAD+ and ATP levels, M1/2 macrophage-related markers, and expression of the SIRT1/NF-κB pathway were detected. RESULTS In MH-S cells, NMN significantly decreased the apoptotic rate from 12.25% to 5.74%. In septic mice, NMN improved the typical pathologic findings in lungs and reduced W/D ratio and MPO activity, but increased NAD+ and ATP levels. Additionally, NMN suppressed M1 but promoted M2 polarization, and upregulated the expression of SIRT1, with inhibition of NF-κB-p65 acetylation and phosphorylation. Furthermore, inhibition of SIRT1 reversed the effects of NMN-induced M2 macrophage polarization. CONCLUSIONS NMN protects against sepsis-induced ALI by promoting M2 macrophage polarization via the SIRT1/NF-κB pathway, it might be an effective strategy for preventing or treating sepsis-induced ALI.
Collapse
Affiliation(s)
- Simeng He
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Xianhong Jiang
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Jing Yang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases of Integrated Traditional Chinese and Western Medicine, Tianjin Nankai Hospital, Tianjin, China
| | - Ya Wu
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Jia Shi
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Xiaoyang Wu
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Shihan Du
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Yuan Zhang
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Lirong Gong
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Shuan Dong
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Jianbo Yu
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
180
|
Zheng Q, Xing J, Li X, Tang X, Zhang D. PRDM16 suppresses ferroptosis to protect against sepsis-associated acute kidney injury by targeting the NRF2/GPX4 axis. Redox Biol 2024; 78:103417. [PMID: 39549609 PMCID: PMC11612791 DOI: 10.1016/j.redox.2024.103417] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 11/02/2024] [Indexed: 11/18/2024] Open
Abstract
Acute kidney injury (AKI) constitutes a significant public health issue. Sepsis accounts for over 50 % of AKI cases in the ICU. Recent findings from our research indicated that the PRD1-BF1-RIZ1 homeodomain protein 16 (PRDM16) inhibited the progression of diabetic kidney disease (DKD). However, its precise role and regulatory mechanism in sepsis-induced AKI remain obscure. This study reveals that lipopolysaccharide (LPS) and cecum ligation and puncture (CLP) instigated PRDM16 expression in Boston University mouse proximal tubule (BUMPT) cells and mouse kidneys, respectively. Functionally, PRDM16 curtailed LPS-induced ferroptosis. Mechanistically, PRDM16 associates with the promoter regions of nuclear factor-erythroid 2-related factor-2 (NRF2) and augments its expression, subsequently enhancing glutathione peroxidase 4 (GPX4) expression. Additionally, PRDM16 directly engages with the promoter regions of GPX4, stimulating its expression. Notably, these observations were corroborated in human renal tubular epithelial (HK-2) cells. Furthermore, the ablation of PRDM16 from kidney proximal tubules in mice inhibited NRF2 and GPX4 expression, leading to decreased glutathione (GSH)/oxidized glutathione (GSSG) ratio, increased Fe2+ and reactive oxygen species (ROS) production, exacerbated ferroptosis, and AKI progression. Conversely, PRDM16 knock-in exhibited the opposite effects. Ultimately, adenovirus (ADV)-PRDM16 plasmid or poly (lactide-glycolide acid) (PLGA)-encapsulated formononetin not only mitigated sepsis-induced AKI but also alleviated liver, cardiac, and lung injury. In summary, PRDM16 inhibits ferroptosis via the NRF2/GPX4 axis or GPX4 to prevent sepsis-induced multi-organ injury, including AKI. PLGA-encapsulated formononetin presents a promising therapeutic approach.
Collapse
Affiliation(s)
- Qiang Zheng
- Department of Emergency, The First Hospital of Jilin University, Changchun, Jilin, China; Department of Emergency and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jihong Xing
- Department of Emergency, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xiaozhou Li
- Department of Emergency and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xianming Tang
- Department of Emergency and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Dongshan Zhang
- Department of Emergency and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China; Emergency Medicine and Difficult Diseases Institute, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China; Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China; Furong Laboratory, Changsha, Hunan, China.
| |
Collapse
|
181
|
Wang ZX, Qin RJ, Yu LL, Nurlan A, Jing XH, Li M. Acupuncture Treats Sepsis through Immune Modulation and Organ Protection. Curr Med Sci 2024; 44:1185-1192. [PMID: 39673001 DOI: 10.1007/s11596-024-2957-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/14/2024] [Indexed: 12/15/2024]
Abstract
Sepsis is a secondary condition resulting from severe systemic infections. It is a significant contributor to mortality in critically ill patients with rapid onset and severe symptoms. Acupuncture is a traditional Chinese medical treatment. Recent clinical studies have demonstrated that acupuncture, as an important synergistic therapy, has promising therapeutic effects in the treatment of sepsis. This paper reviews the mechanisms of immunomodulation and target organ protection associated with acupuncture and synergistic drug acupuncture in the treatment of sepsis. It also integrates existing studies to elucidate the modulation of the immune system and the protective effect of acupuncture on target organs.
Collapse
Affiliation(s)
- Zi-Xiao Wang
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ren-Jie Qin
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ling-Ling Yu
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Arman Nurlan
- College of Acupuncture and Massage, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330000, China
| | - Xiang-Hong Jing
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100000, China.
| | - Man Li
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
182
|
Chen Y, Fan C, Wang J, Jiang M. Rivaroxaban Combined with Atorvastatin Inhibits Acute Pulmonary Embolism by Promoting the Expression of NRF2/NQO1. Cardiovasc Drugs Ther 2024; 38:1271-1287. [PMID: 37316694 DOI: 10.1007/s10557-023-07479-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/02/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND Atorvastatin and direct oral factor Xa inhibitors (for instance, rivaroxaban) are co-administrated in patients with atrial fibrillation. However, no studies have been conducted on the function of these two agents in acute pulmonary embolism (APE). Therefore, we investigated the effects of rivaroxaban + atorvastatin in rats with APE and explored the underlying mechanisms. METHODS Patients with APE were enrolled, and rats with APE were generated for different regimens. The mean pulmonary arterial pressure (mPAP), heart rate, and PaO2 of APE patients and rats were measured. The plasma levels of oxidative stress- and inflammation-related factors were measured, and the expression of platelet activation markers (CD63 and CD62P) was detected. The proteins targeted by rivaroxaban and atorvastatin, the targets associated with APE, and the genes aberrantly expressed in rats with APE were intersected to obtain candidate factors. RESULTS Rivaroxaban + atorvastatin reduced mPAP and increased PaO2 in patients and rats with APE. Rivaroxaban + atorvastatin repressed oxidative stress, inflammatory levels, and platelet activation during APE. NRF2 and NQO1 were increased in the lung of rats treated with rivaroxaban + atorvastatin. The therapeutic effect of the combination on APE rats was suppressed after NRF2 downregulation. NRF2 promoted the NQO1 transcription. NQO1 eliminated the inhibitory effect of sh-NRF2 on the combined therapy. CONCLUSION The alleviating effect of rivaroxaban + atorvastatin administration against APE correlates with NRF2/NQO1 expression.
Collapse
Affiliation(s)
- Yang Chen
- Department of Respiratory and Critical Care Medicine, Cangzhou Central Hospital, No. 16, Xinhua West Road, Yunhe District, Cangzhou, 061000, Hebei, P.R. China
| | - Cuncun Fan
- Department of Neurosurgery, Cangzhou Central Hospital, Cangzhou, 061000, Hebei, P.R. China
| | - Jinkun Wang
- Department of Respiratory and Critical Care Medicine, Cangzhou Central Hospital, No. 16, Xinhua West Road, Yunhe District, Cangzhou, 061000, Hebei, P.R. China
| | - Mingming Jiang
- Department of Respiratory and Critical Care Medicine, Cangzhou Central Hospital, No. 16, Xinhua West Road, Yunhe District, Cangzhou, 061000, Hebei, P.R. China.
| |
Collapse
|
183
|
Li R, Strobl J, Poyner EFM, Balbaa A, Torabi F, Mazin PV, Chipampe NJ, Stephenson E, Ramírez-Suástegi C, Shanmugiah VBM, Gardner L, Olabi B, Coulthard R, Botting RA, Zila N, Prigmore E, Gopee NH, Chroscik MA, Kritikaki E, Engelbert J, Goh I, Chan HM, Johnson HF, Ellis J, Rowe V, Tun W, Reynolds G, Yang D, Foster AR, Gambardella L, Winheim E, Admane C, Rumney B, Steele L, Jardine L, Nenonen J, Pickard K, Lumley J, Hampton P, Hu S, Liu F, Liu X, Horsfall D, Basurto-Lozada D, Grimble L, Bacon CM, Weatherhead SC, Brauner H, Wang Y, Bai F, Reynolds NJ, Allen JE, Jonak C, Brunner PM, Teichmann SA, Haniffa M. Cutaneous T cell lymphoma atlas reveals malignant T H2 cells supported by a B cell-rich tumor microenvironment. Nat Immunol 2024; 25:2320-2330. [PMID: 39558094 PMCID: PMC11588665 DOI: 10.1038/s41590-024-02018-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 10/11/2024] [Indexed: 11/20/2024]
Abstract
Cutaneous T cell lymphoma (CTCL) is a potentially fatal clonal malignancy of T cells primarily affecting the skin. The most common form of CTCL, mycosis fungoides, can be difficult to diagnose, resulting in treatment delay. We performed single-cell and spatial transcriptomics analysis of skin from patients with mycosis fungoides-type CTCL and an integrated comparative analysis with human skin cell atlas datasets from healthy and inflamed skin. We revealed the co-optation of T helper 2 (TH2) cell-immune gene programs by malignant CTCL cells and modeling of the tumor microenvironment to support their survival. We identified MHC-II+ fibroblasts and dendritic cells that can maintain TH2 cell-like tumor cells. CTCL tumor cells are spatially associated with B cells, forming tertiary lymphoid structure-like aggregates. Finally, we validated the enrichment of B cells in CTCL and its association with disease progression across three independent patient cohorts. Our findings provide diagnostic aids, potential biomarkers for disease staging and therapeutic strategies for CTCL.
Collapse
Affiliation(s)
- Ruoyan Li
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK.
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
| | - Johanna Strobl
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Elizabeth F M Poyner
- Biosciences Institute, Newcastle University, Newcastle, UK
- Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle, Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Aya Balbaa
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | - Pavel V Mazin
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | - Emily Stephenson
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Biosciences Institute, Newcastle University, Newcastle, UK
| | | | | | - Louis Gardner
- Biosciences Institute, Newcastle University, Newcastle, UK
- Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle, Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Bayanne Olabi
- Biosciences Institute, Newcastle University, Newcastle, UK
- Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle, Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Rowen Coulthard
- NovoPath, Department of Cellular Pathology, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Rachel A Botting
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Nina Zila
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
- Section Biomedical Science, University of Applied Sciences FH Campus Wien, Vienna, Austria
| | - Elena Prigmore
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Nusayhah H Gopee
- Biosciences Institute, Newcastle University, Newcastle, UK
- Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle, Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Marta A Chroscik
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Efpraxia Kritikaki
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Justin Engelbert
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Issac Goh
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Hon Man Chan
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | - Jasmine Ellis
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Victoria Rowe
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Win Tun
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Gary Reynolds
- Biosciences Institute, Newcastle University, Newcastle, UK
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA
| | - Dexin Yang
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | | | | | - Elena Winheim
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Chloe Admane
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Benjamin Rumney
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Lloyd Steele
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Laura Jardine
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Julia Nenonen
- Division of Dermatology, Department of Medicine, Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Keir Pickard
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Jennifer Lumley
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Philip Hampton
- Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle, Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Simeng Hu
- Biomedical Pioneering Innovation Center and School of Life Sciences, Peking University, Beijing, China
| | - Fengjie Liu
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China
| | - Xiangjun Liu
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China
| | - David Horsfall
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Daniela Basurto-Lozada
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Louise Grimble
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Chris M Bacon
- Wolfson Childhood Cancer Research Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Department of Cellular Pathology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Sophie C Weatherhead
- Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle, Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Hanna Brauner
- Division of Dermatology, Department of Medicine, Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Dermatology, Karolinska University Hospital, Stockholm, Sweden
| | - Yang Wang
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China
| | - Fan Bai
- Biomedical Pioneering Innovation Center and School of Life Sciences, Peking University, Beijing, China
| | - Nick J Reynolds
- Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle, Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Judith E Allen
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Constanze Jonak
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Patrick M Brunner
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK.
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK.
- Department of Medicine, University of Cambridge, Cambridge, UK.
| | - Muzlifah Haniffa
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK.
- Biosciences Institute, Newcastle University, Newcastle, UK.
- Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle, Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.
| |
Collapse
|
184
|
Tang X, Zhu Y, Cao Z, Wang X, Cai X, Tang Y, Zhou J, Wu M, Zhen X, Ding L, Yan G, Wang H, Sun H, Jiang R. CDC42 deficiency leads to endometrial stromal cell senescence in recurrent implantation failure. Hum Reprod 2024; 39:2768-2784. [PMID: 39487595 PMCID: PMC11630066 DOI: 10.1093/humrep/deae246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 09/22/2024] [Indexed: 11/04/2024] Open
Abstract
STUDY QUESTION Does the downregulation of cell division cycle 42 (CDC42) protein in endometrial stroma lead to endometrial senescence in patients with recurrent implantation failure (RIF), and what is the potential mechanism? SUMMARY ANSWER CDC42 deficiency causes endometrial stromal senescence and decidualization defects, impairing uterine receptivity of RIF patients, via activation of Wnt signaling pathway. WHAT IS KNOWN ALREADY Uterine aging is unique due to the cyclic remodeling and decidualization of endometrial tissue. Several transcriptomic studies have reported increased senescence in the endometrium in young patients with RIF. Our previous transcriptomic sequencing study discovered that endometrium from women with RIF showed downregulation of CDC42, which is an essential molecule affected by various senescence-related diseases. STUDY DESIGN, SIZE, DURATION The endometrial samples of a total of 71 fertile control patients and 37 RIF patients were collected to verify the association between CDC42 expression and endometrial senescence of RIF patients. Primary endometrial stromal cells (EnSCs) were isolated from endometrial biopsies taken from patients without any endometrial complications and planning to undergo IVF, then subjected to adenovirus-mediated CDC42 knockdown and decidualization induction to explore the detailed mechanism by which CDC42 governs stromal senescence and decidualization. Wnt inhibitor XAV-939 was used to correct the endometrial senescence and decidualization defect. PARTICIPANTS/MATERIALS, SETTING, METHODS Senescence was determined by cell cycle arrest markers (e.g. P16, P21, and P53), SASP molecules (e.g. IL6 and CXCL8), and SA-β-gal staining. Masson's staining and Sirius Red staining were used to detect the endometrial fibrosis. Decidualization was evaluated by the mRNA expression and protein secretion of PRL and IGFBP1, F-actin immunostaining, and the BeWo spheroids 'in vitro implantation' model. Methods used to assess cell function included adenovirus transduction, RNA-sequencing, bioinformatic analysis, western blotting, RT-qPCR, ELISA, and immunofluorescence. MAIN RESULTS AND THE ROLE OF CHANCE Here, we observed remarkably increased levels of stromal senescence and fibrosis, along with stromal CDC42 deficiency, in the endometrium of patients with RIF (P < 0.001). Knockdown of CDC42 effectively induced premature senescence in EnSCs, leading to aberrant accumulation of senescent EnSCs and collagen deposition during decidualization. CDC42 deficiency in EnSCs restrained the decidualization differentiation and receptivity to trophoblast cells. Transcriptomic analysis revealed Wnt signaling activation as a critical downstream alteration in CDC42-deficient EnSCs. Mechanistically, CDC42 interacted with AKT competitively to impede the binding of GSK3β to AKT. Knockdown of CDC42 increased AKT-mediated phosphorylation of GSK3β to inactivate the Axin-GSK3β destruction complex, leading to accumulation and nuclear translocation of β-catenin. Importantly, Wnt signaling inhibitors partially corrected the endometrial senescence caused by CDC42 deficiency, and improved both decidualization and trophoblast invasion. LARGE SCALE DATA RNA-seq data sets generated in this study have been deposited at the NCBI database with BioProject accession number PRJNA1102745. LIMITATIONS, REASONS FOR CAUTION The present study was based on in vitro cell cultures. Further studies involving CDC42-regulated endometrial senescence are needed in knockout mice model and human endometrial assembloids. WIDER IMPLICATIONS OF THE FINDINGS In addition to uncovering endometrial senescence in RIF, our findings underscore the significance of CDC42 in modulating EnSC senescence to maintain the decidualization function, and suggest Wnt signaling inhibitors as potential therapeutic agents for alleviating endometrial senescence. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the National Natural Science Foundation of China [82271698 (R.J.), 82030040 (H.S.), 82288102 (H.W.), and 82371680 (G.Y.)]; the Natural Science Foundation of Jiangsu Province [BK20231117 (R.J.)]; and the Medical Science and Technology Development Foundation of Nanjing Department of Health [YKK23097 (Y.Z.)]. The authors declare no potential conflicts of interest.
Collapse
Affiliation(s)
- Xinyi Tang
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Yingchun Zhu
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Zhiwen Cao
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Xiaoying Wang
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Xinyu Cai
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Yurun Tang
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Jidong Zhou
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Min Wu
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Xin Zhen
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Lijun Ding
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Guijun Yan
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Haibin Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Haixiang Sun
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
| | - Ruiwei Jiang
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| |
Collapse
|
185
|
Wang Z, Guo Y, Li K, Huo Y, Wang S, Dong S, Ma M. Targeting the PI3K/mTOR pathway in idiopathic pulmonary fibrosis: Advances and therapeutic potential. Bioorg Med Chem 2024; 115:117908. [PMID: 39471771 DOI: 10.1016/j.bmc.2024.117908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 11/01/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive, fatal lung disease characterized by irreversible tissue scarring, leading to severe respiratory dysfunction. Despite current treatments with the drugs Pirfenidone and Nintedanib, effective management of IPF remains inadequate due to limited therapeutic benefits and significant side effects. This review focuses on the phosphoinositide 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) signaling pathway, a critical regulator of cellular processes linked to fibrosis, such as fibroblast proliferation, inflammation, and epithelial-mesenchymal transition (EMT). We discuss recent advances in understanding the role of the PI3K/mTOR pathway in IPF pathogenesis and highlight emerging therapies targeting this pathway. The review compiles evidence from both preclinical and clinical studies, suggesting that PI3K/mTOR inhibitors may offer new hope for IPF treatment by modulating fibrosis and improving patient outcomes. Moreover, it outlines the potential for these inhibitors to be developed into effective, personalized treatment options, underscoring the importance of further research to explore their efficacy and safety profiles comprehensively.
Collapse
Affiliation(s)
- Zhengyang Wang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Yanzhi Guo
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Kaiyin Li
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Yan Huo
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Shuyan Wang
- Department of Anesthesiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Suzhen Dong
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China.
| | - Mingliang Ma
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China; Wenzhou Key Laboratory of Biophysics, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
186
|
Duan X, Hu T, Xu L, Li Z, Jing J, Xu D, Ding J, Li F, Jiang M, Wang J. The Correlation Analysis Between m6A Methylation Modification and Ferroptosis Induced by Cigarette Smoke in Human Bronchial Epithelium. Immun Inflamm Dis 2024; 12:e70104. [PMID: 39688470 PMCID: PMC11650891 DOI: 10.1002/iid3.70104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/11/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD), a prevalent respiratory condition, is characterized by long-term airway inflammation, which can lead to airway remodeling and persistent airflow restriction. Exposure to cigarette smoke is known as a major contributor to COPD development. Research has confirmed that ferroptosis and m6A modification are closely related to various inflammatory-related diseases. However, the correlation between m6A methylation and ferroptosis in COPD has not been confirmed. In this study, combined with bioinformatics analysis and molecular biology methods we investigated how m6A methylation was correlated to ferroptosis-associated genes (SLC7A11 and NQO-1) in cigarette smoke induced 16HBES cells. METHODS Two microarray datasets (GSE30063 and GSE64614) were combined to identify differentially expressed genes (DEGs) through the application of bioinformatics techniques. A cigarette smoke (CS)-induced 16HBE cells model was established. The ROS, GSH, MDA, and total iron content were detected by relevant detection kits. The expression levels associated with ferroptosis and m6A methylation modification-related genes were determined via reverse transcription-quantitative polymerase chain reaction and western blot. RESULTS Overall, 529 DEGs were identified in the above two databases. For COPD patients, significant changes were observed in FAGs (GCLC, NQO-1, SLC7A11) and m6A methylation-related genes (FTO). A negative correlation was also noted between the expression level of genes linked to ferroptosis (SLC7A11 and NQO-1) and that of the m6A methylation gene (FTO). The in vitro experiments results indicate that SLC7A11 and NQO-1 were significantly downregulated, and FTO were significantly upregulated. In addition, cigarette smoke stimulation increased the levels of MDA, LPO, and ROS, while reducing the content of GSH and total iron content in 16HBE cells. CONCLUSION Our findings explored the relationship between ferroptosis and m6A methylation in COPD, and screened out SLC7A11, NQO-1 and FTO may be critical in the pathogenesis of COPD.
Collapse
Affiliation(s)
- Xiaomei Duan
- Department of Xinjiang Laboratory of Respiratory Disease ResearchTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
- Department of Xinjiang Clinical Research Center for Respiratory DiseasesTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
| | - Tingting Hu
- Department of Xinjiang Laboratory of Respiratory Disease ResearchTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
- Department of Xinjiang Clinical Research Center for Respiratory DiseasesTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
| | - Lijuan Xu
- Department of EndocrineTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
| | - Zheng Li
- Department of Xinjiang Laboratory of Respiratory Disease ResearchTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
- Department of Xinjiang Clinical Research Center for Respiratory DiseasesTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
| | - Jing Jing
- Department of Xinjiang Laboratory of Respiratory Disease ResearchTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
- Department of Xinjiang Clinical Research Center for Respiratory DiseasesTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
| | - Dan Xu
- Department of Xinjiang Laboratory of Respiratory Disease ResearchTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
- Department of Xinjiang Clinical Research Center for Respiratory DiseasesTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
| | - Jianbing Ding
- Department of Immunology, College of Basic MedicineXinjiang Medical UniversityUrumqiXinjiangChina
| | - Fengsen Li
- Department of Xinjiang Laboratory of Respiratory Disease ResearchTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
- Department of Xinjiang Clinical Research Center for Respiratory DiseasesTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
| | - Min Jiang
- Department of Xinjiang Laboratory of Respiratory Disease ResearchTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
- Department of Xinjiang Clinical Research Center for Respiratory DiseasesTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
| | - Jing Wang
- Department of Xinjiang Laboratory of Respiratory Disease ResearchTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
- Department of Xinjiang Clinical Research Center for Respiratory DiseasesTraditional Chinese Medicine Hospital Affiliated to Xinjiang Medical UniversityUrumqiXinjiangChina
| |
Collapse
|
187
|
Shi J, Piao M, Liu C, Yang J, Guan X, Liu H, Li Q, Zhang Y, Yu J. Electroacupuncture pretreatment maintains mitochondrial quality control via HO-1/MIC60 signaling pathway to alleviate endotoxin-induced acute lung injury. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167480. [PMID: 39209235 DOI: 10.1016/j.bbadis.2024.167480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/04/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Electroacupuncture has been demonstrated to mitigate endotoxin-induced acute lung injury by enhancing mitochondrial function. This study investigates whether electroacupuncture confers lung protection through the regulation of mitochondrial quality control mediated by heme oxygenase-1 (HO-1) and the mitochondrial inner membrane protein MIC60. HO-1, an inducible stress protein, is crucial for maintaining mitochondrial homeostasis and protecting against lung injury. MIC60, a key component of the mitochondrial contact site and cristae organizing system, supports mitochondrial integrity. We employed genetic knockout/silencing and cell transfection techniques to model lipopolysaccharide (LPS)-induced lung injury, assessing changes in mitochondrial structure, reactive oxygen species (ROS) production, mitochondrial membrane potential (MMP), and the expression of proteins essential for mitochondrial quality control. Our findings reveal that electroacupuncture alleviates endotoxin-induced acute lung injury and associated mitochondrial dysfunction, as evidenced by reductions in lung injury scores, decreased ROS production, and suppressed expression of proteins involved in mitochondrial fission and mitophagy. Additionally, electroacupuncture enhanced MMP and upregulated proteins that facilitate mitochondrial fusion and biogenesis. Importantly, the protective effects of electroacupuncture were reduced in models with Hmox1 knockout or Mic60 silencing, and in macrophages transfected with Hmox1-siRNA or Mic60-siRNA. Moreover, HO-1 was found to influence MIC60 expression during electroacupuncture preconditioning and LPS challenge, demonstrating that these proteins not only co-localize but also interact directly. In conclusion, electroacupuncture effectively modulates mitochondrial quality control through the HO-1/MIC60 signaling pathway, offering an adjunctive therapeutic strategy to ameliorate endotoxin-induced acute lung injury in both in vivo and in vitro settings.
Collapse
Affiliation(s)
- Jia Shi
- Tianjin Nankai Hospital, Tianjin Medical University; Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin; Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair
| | - Meiling Piao
- Tianjin Nankai Hospital, Tianjin Medical University; Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin; Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair
| | - Chuanning Liu
- Tianjin Nankai Hospital, Tianjin Medical University; Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin; Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair
| | - Jing Yang
- Tianjin Nankai Hospital, Tianjin Medical University; Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin; Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair
| | - Xin Guan
- Tianjin Nankai Hospital, Tianjin Medical University; Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin; Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair
| | - Huayang Liu
- Tianjin Nankai Hospital, Tianjin Medical University; Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin; Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair
| | - Qiujia Li
- Tianjin Nankai Hospital, Tianjin Medical University; Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin; Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair
| | - Yuan Zhang
- Tianjin Nankai Hospital, Tianjin Medical University; Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin; Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair
| | - Jianbo Yu
- Tianjin Nankai Hospital, Tianjin Medical University; Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin; Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair.
| |
Collapse
|
188
|
Cheng D, Lian W, Jia X, Wang T, Sun W, Liu Y, Ni C. LGALS3 regulates endothelial-to-mesenchymal transition via PI3K/AKT signaling pathway in silica-induced pulmonary fibrosis. Toxicology 2024; 509:153962. [PMID: 39353502 DOI: 10.1016/j.tox.2024.153962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/27/2024] [Accepted: 09/28/2024] [Indexed: 10/04/2024]
Abstract
Silicosis is a progressive and chronic occupational lung disease characterized by lung inflammation, silicotic nodule formation, and diffuse pulmonary fibrosis. Emerging evidence indicates that endothelial-mesenchymal transition (EndoMT) plays a crucial role in the development of silicosis. Herein, we conducted a SiO2-induced EndoMT model and established a mouse model with pulmonary fibrosis by silica. We identified that SiO2 effectively increased the expression of mesenchymal markers while decreasing the levels of endothelial markers in endothelial cells. It's further demonstrated that SiO2 induced the PI3K/Akt signaling pathway activation via LGALS3 synthesis. Next, interfering LGALS3 blocked the process of EndoMT by inhibiting the activity of PI3K/AKT signaling. In vivo, the administration of a specific PI3K inhibitor LY294002 significantly alleviated silica-induced pulmonary fibrosis. Collectively, these results identified that the LGALS3/PI3K/AKT pathway provided a rationale target for the clinical treatment and intervention of silicosis.
Collapse
Affiliation(s)
- Demin Cheng
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China; Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Wenxiu Lian
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Xinying Jia
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Ting Wang
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210000, China
| | - Wenqing Sun
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yi Liu
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China.
| | - Chunhui Ni
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Department of Public Health, Kangda College of Nanjing Medical University, Lianyungang 320700, China.
| |
Collapse
|
189
|
Sun L, He X, Kong J, Zhou J. Protection of Qingfei Xieding prescription from idiopathic pulmonary fibrosis by regulating renin-angiotensin and ferroptosis in MLE-12 cells. Histol Histopathol 2024; 39:1643-1658. [PMID: 38666295 DOI: 10.14670/hh-18-746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lifelong lung disease, but there is no specific drug for treatment. Qingfei Xieding prescription (QF) is active in the treatment of lung diseases. More comprehensive mechanisms over how QF exhibits anti-pulmonary fibrosis need to be elucidated. TGF-β was used to construct a pulmonary fibrosis cell model in vitro. Bleomycin was applied to induce a lung tissue fibrosis model in mice in vivo. Flow cytometry was used to detect cellular ROS and lipid oxidation levels. Cell substructure was observed by Transmission Electron Microscopy. ELISA was used to determine the levels of inflammatory factors. HE staining, Masson staining and immunohistochemistry were performed to evaluate the degree of fibrosis. Western Blot assay was used to determine the protein expressions of different molecules. In TGF-β-exposed lung epithelial MLE-12 cell model, α-SMA and Collagen I were significantly elevated and cell viability was reduced. QF treatment restored the cell viability decreased by exogenous TGF-β. Ferroptosis inducer Erastin administration could reverse the beneficial effects such as lipid oxidation and ROS reduction caused by QF treatment. QF was proven to inhibit ferroptosis and alleviated the process of IPF by activating ACE2 signal axis. In bleomycin induced IPF mice model, QF altered lung coefficient, body weight and the expression of inflammatory factors, which were prevented by ferroptosis activator Erastin. QF was demonstrated to affect the ACE2-ERK signaling axis in vivo. QF alleviated idiopathic pulmonary fibrosis by regulating renin-angiotensin through blocking ferroptosis. This research offers evidence for the potentiality of QF in clinical application for IPF therapy.
Collapse
Affiliation(s)
- Lifang Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
- Department of Tuberculosis, Hangzhou Red Cross Hospital, Hangzhou, PR China
| | - Xinxin He
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou, PR China
| | - Jiao Kong
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Jianying Zhou
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.
| |
Collapse
|
190
|
Fei Q, Zhang J, Chen L, Shi M, Wang Q, Xu F, Shi J, Qin Y. Modulating ferroptosis and mycobactericidal activity in lung epithelial cells via YY1/iNOS pathway. Life Sci 2024; 358:123131. [PMID: 39424267 DOI: 10.1016/j.lfs.2024.123131] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 09/30/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Mycobacterium tuberculosis infection triggers various forms of host cell death, including ferroptosis in lung epithelial cells; YY1, a critical transcription factor, plays a pivotal role in regulating ferroptosis, however, the underlying mechanisms are not fully understood. METHODS To investigate Mycobacterium marinum (M.marinum) infection in lung epithelial cells A549 and H1299, we utilized flow cytometry to evaluate cell death and measure reactive oxygen species (ROS). Colony-forming unit (CFU) assays determined the intracellular bacterial load. Ferroptosis was analyzed using a specific detection kit to measure malondialdehyde (MDA) and glutathione (GSH) levels. The interaction between the transcription factor YY1 and the iNOS promoter was assessed through a dual-luciferase reporter assay. RESULTS M.marinum induced ferroptosis in lung epithelial cells through invasion. This effect is most pronounced at 8 h of infection and decreases over time but increased with a higher multiplicity of infection (MOI). YY1 knockdown decreases the expression of SLC7A11 and GPX4, attenuates cellular ferroptosis, while YY1 overexpression has the opposite phenomenon, enhancing the expression of bactericidal molecules such as iNOS and MPEG1, thereby markedly reducing the intracellular bacterial load. We identified substantial binding of YY1 to the iNOS promoter region (-655 to -1018 bp), enhancing mycobactericidal activity in YY1-overexpressing cells. CONCLUSIONS Our study demonstrates that YY1 inhibits ferroptosis induced by Mycobacterium marinum infection and reduces intracellular bacterial proliferation in lung epithelial cells. These findings provide a crucial basis for developing anti-tuberculosis therapies that target YY1 modulation, potentially offering new clinical avenues for the treatment of tuberculosis.
Collapse
Affiliation(s)
- Qiuwen Fei
- Department of Pathogen Biology, Medical College, Nantong University, No. 19 Qixiu Road, Nantong, China
| | - Jian Zhang
- Affiliated Haian Hospital of Nantong University, Haian, China
| | - Liangqiong Chen
- Affiliated Haian Hospital of Nantong University, Haian, China
| | - Manqi Shi
- Department of Pathogen Biology, Medical College, Nantong University, No. 19 Qixiu Road, Nantong, China
| | - Qinglan Wang
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Feifan Xu
- Department of Clinical Laboratory, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Nantong, China
| | - Jiahai Shi
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, Nantong Clinical Medical Research Center of Cardiothoracic Disease, Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, China
| | - Yongwei Qin
- Department of Pathogen Biology, Medical College, Nantong University, No. 19 Qixiu Road, Nantong, China.
| |
Collapse
|
191
|
Li Y, Li M, Mao J, Guo Q, Zhu W, Fu R, Wan X, Dong W, Li L, Mao C, Ji D, Zhang K, Lu T. The processing mechanism of vinegar-processed Curcumae Rhizome enhances anti hepatic fibrotic effects through regulation of PI3K/Akt/mTOR signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156098. [PMID: 39395324 DOI: 10.1016/j.phymed.2024.156098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/15/2024] [Accepted: 09/26/2024] [Indexed: 10/14/2024]
Abstract
BACKGROUND Hepatic fibrosis, a chronic pathological condition resulting from various forms of persistent liver injury, in the later stage, it can evolve into cirrhosis and even liver cancer. Curcumae Rhizoma (CR), traditionally recognized for its properties in line qi break blood, eliminate accumulation and relieve pain. According to traditional Chinese medicine (TCM) principles, vinegar-processing enhances CR's ability to enter the liver meridian and act on the blood level, potentially augmenting its therapeutic effects on hepatic diseases. Therefore, vinegar-processed Curcumae Rhizoma (VCR) is frequently employed in treating liver fibrosis and related hepatic conditions. However, the underlying mechanisms of vinegar processing in enhancing its therapeutic efficacy remain unclear. METHODS The anti-liver fibrosis effects of CR and VCR were verified at individual and cellular levels. Subsequently, HPLC-Q-TOFMS and pharmacokinetic analysis were utilized to elucidate the potential bioactive substances underlying the enhanced anti-fibrotic efficacy of VCR. Building upon these findings, network pharmacology and metabolomics were integrated to screen for key effect components and regulatory pathways. Finally, the mechanisms of action were further analyzed and validated at the tissue and cellular levels through Western blotting (WB) and molecular docking studies. RESULTS Both CR and VCR exhibited therapeutic effects against hepatic fibrosis, with VCR demonstrating enhanced efficacy after vinegar processing. 6 sesquiterpenes including furanodiene and curdione, showed significant alterations in plasma exposure and hepatic distribution post-processing. VCR significantly improved pathological liver conditions, lipid accumulation, and fibrosis severity. Additionally, VCR markedly reduced the expression of α-SMA in the liver and attenuated the elevations in liver function markers such as ALT and AST. Combined network pharmacology, metabolomics, and hepatic tissue WB analysis revealed that the reduced phosphorylation of the PI3K/Akt/mTOR pathway is a critical mechanism in VCR's anti-fibrotic effects. Experiments on LX-2 cells demonstrated that four sesquiterpenes, including furanodiene and curdione, effectively inhibited the proliferation of activated hepatic stellate cells (HSCs). Furanodiene, in particular, promoted apoptosis in activated HSCs by reducing phosphorylation levels of the PI3K/Akt/mTOR pathway proteins, increasing BAX expression, and activating downstream caspase-3 to achieve the effect of anti-liver fibrosis. CONCLUSION Vinegar-processing significantly increases the plasma exposure and hepatic distribution of components such as furanodiene in VCR, enhancing anti-fibrotic efficacy by downregulating the phosphorylation levels of the PI3K/Akt/mTOR pathway and promoting HSC apoptosis. This study provides a comprehensive explanation of the vinegar-processing mechanism and its role in enhancing the anti-fibrotic effects of VCR, offering insights for its clinical application in liver fibrosis treatment and reference for the mechanistic study of other vinegar-processed herbal medicines.
Collapse
Affiliation(s)
- Yu Li
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Mingxuan Li
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jing Mao
- College of Medical, Nanjing University of Chinese Medicine, Nanjing, 210023, China; The First Clinical School of Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qiang Guo
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wenhong Zhu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Rao Fu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xin Wan
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wenhao Dong
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lin Li
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chunqin Mao
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - De Ji
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Kewei Zhang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Tulin Lu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
192
|
Yao C, Li Z, Su H, Sun K, Liu Q, Zhang Y, Zhu L, Jiang F, Fan Y, Shou S, Wu H, Jin H. Integrin subunit beta 6 is a potential diagnostic marker for acute kidney injury in patients with diabetic kidney disease: a single cell sequencing data analysis. Ren Fail 2024; 46:2409348. [PMID: 39356055 PMCID: PMC11448326 DOI: 10.1080/0886022x.2024.2409348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Diabetic kidney disease (DKD), a prevalent complication of diabetes mellitus, is often associated with acute kidney injury (AKI). Thus, the development of preventive and therapeutic strategies is crucial for delaying the progression of AKI and DKD. METHODS The GSE183276 dataset, comprising the data of 20 healthy controls and 12 patients with AKI, was downloaded from the Gene Expression Omnibus (GEO) database to analyze the AKI group. For analyzing the DKD group, the GSE131822 dataset, comprising the data of 3 healthy controls and 3 patients with DKD, was downloaded from the GEO database. The common differentially expressed genes (DEGs) in renal tubular epithelial cells (TECs) were subjected to enrichment analyses. Next, a protein-protein interaction (PPI) network was constructed using the Search Tool for the Retrieval of Interacting Genes database to analyze gene-related regulatory networks. Finally, the AKI animal models and the DKD and AKI cell models were established, and the reliability of the identified genes was validated using quantitative real-time polymerase chain reaction analysis. RESULTS Functional analysis was performed with 40 common DEGs in TECs. Eight hub genes were identified using the PPI and gene-related networks. Finally, validation experiments with the in vivo animal model and the in vitro cellular model revealed the four common DEGs. Four DEGs that share molecular mechanisms in the pathogenesis of DKD and AKI were identified. In particular, the expression of Integrin Subunit Beta 6(ITGB6), a hub and commonly upregulated gene, was upregulated in the in vitro models. CONCLUSION ITGB6 may serve as a biomarker for early AKI diagnosis in patients with DKD and as a target for early intervention therapies.
Collapse
Affiliation(s)
- Congcong Yao
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Ziwei Li
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongshuang Su
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Keke Sun
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Qihui Liu
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan Zhang
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Lishuang Zhu
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Feng Jiang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yaguang Fan
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Songtao Shou
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Heng Wu
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Heng Jin
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
193
|
Zhang WJ, Chen D. Mesenchymal stem cell transplantation plays a role in relieving cancer pain. Front Pharmacol 2024; 15:1483716. [PMID: 39679363 PMCID: PMC11637888 DOI: 10.3389/fphar.2024.1483716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024] Open
Abstract
Tumors can invade, compress, and damage nerves, leading to persistent pain and seriously affecting the quality of life of patients. However, their treatment is challenging. Sensitization of peripheral receptors, abnormal activity of primary sensory neurons, activation of glial cells, enhanced inflammatory responses, and sensory information transmission contribute towards cancer pain. Therefore, considerable attention has been paid to exploring prospective methods to inhibit the occurrence of these factors and relieve cancer pain. Studies on different types of pains have revealed that the transplantation of functionally active cells into the host has the pharmacological effect of producing analgesia. Mesenchymal stem cells (MSCs) can act as small active pumps to reduce the expression of pain-related molecules and produce analgesic effects. Moreover, MSCs can establish complex communication networks with non-tumor and cancer cells in the microenvironment, interact with each other, and can be used as destinations for inflammation and tumor sites, affecting their potential for invasion and metastasis. This emphasizes the key role of MSCs in cancer and pain management. The pain relief mechanisms of MSCs include neuronutrition, neural protection, neural network reconstruction, immune regulation, and improvement of the inflammatory microenvironment around the nerve injury. All of these are beneficial for the recovery of injured or stimulated nerves and the reconstruction of neural function, and play a role in relieving pain. The pain treatment strategy of cell transplantation is to repair injured nerves and produce analgesic pharmacological properties that are different from those of painkillers and other physiotherapies. Although the therapeutic role of MSCs in cancer and pain is in its early stages, the therapeutic value of MSCs for cancer pain has great prospects. Therefore, in this study, we explored the possible mechanism between MSCs and cancer pain, the potential therapeutic role of therapeutic cells in cancer pain, and some problems and challenges.
Collapse
Affiliation(s)
- Wen-Jun Zhang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Nanchang University, Jiangxi Medical college, Nanchang, China
| | - Dingyi Chen
- Emergency department, The Second Affiliated Hospital, Nanchang University, Jiangxi Medical college, Nanchang, China
| |
Collapse
|
194
|
Luo P, Gu Q, Wang J, Li X, Li N, Yang W, Meng X, Zhao M. SS31 alleviates LPS-induced acute lung injury by inhibiting inflammatory responses through the S100A8/NLRP3/GSDMD signaling pathway. Eur J Med Res 2024; 29:567. [PMID: 39609864 PMCID: PMC11605876 DOI: 10.1186/s40001-024-02169-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is an acute, diffuse, inflammatory lung injury caused by various endogenous or exogenous factors. It is currently widely recognized that an excessive inflammatory response resulting from immune imbalance constitutes a crucial pathogenic mechanism in ALI/ARDS. SS31 is a novel mitochondria-targeted antioxidant peptide. This article validates the role of SS31 in lipopolysaccharide (LPS)-induced ALI. METHODS The study applied transcriptome sequencing, immunofluorescence, PCR, immunofluorescence and other methods to explore the mechanism of SS31 in LPS induced ALI. RESULTS Transcriptome sequencing results indicate that LPS-induced ALI is closely associated with immune regulatory processes, the Toll-like receptor pathway, and the NF-κB signaling pathway. The role of SS31 in acute lung injury is closely related to biological processes, such as immune regulation and cell death. This study demonstrated that SS31 can inhibit the expression of inflammatory factors IL-6, IL-1β, IL-18, and TNF-α, and reduce the expression of pyroptosis-related proteins NLRP3, and GSDMD-N. Further analysis revealed that S100A8 may be a key gene in the effect of SS31. LPS stimulation leads to increased expression of S100A8, while SS31 decreases its expression. Recombinant protein S100A8 can attenuate the inhibitory effect of SS31 on IL-1β, IL-18, NLRP3, and GSDMD-N. CONCLUSIONS The research results indicate that SS31 may inhibit the activation of the NLRP3 inflammasome and suppress inflammatory responses by regulating S100A8, thereby alleviating LPS-induced ALI in mice; this process may be related to pyroptosis.
Collapse
Affiliation(s)
- Peiyao Luo
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, No. 2075, Qunli Seventh Avenue, Daoli District, Harbin, 150001, Heilongjiang Province, China
| | - Quankuan Gu
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, No. 2075, Qunli Seventh Avenue, Daoli District, Harbin, 150001, Heilongjiang Province, China
| | - Jianpeng Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, No. 2075, Qunli Seventh Avenue, Daoli District, Harbin, 150001, Heilongjiang Province, China
| | - Xianyong Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, No. 2075, Qunli Seventh Avenue, Daoli District, Harbin, 150001, Heilongjiang Province, China
| | - Nana Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, No. 2075, Qunli Seventh Avenue, Daoli District, Harbin, 150001, Heilongjiang Province, China
| | - Wei Yang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, No. 2075, Qunli Seventh Avenue, Daoli District, Harbin, 150001, Heilongjiang Province, China.
| | - Xianglin Meng
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, No. 2075, Qunli Seventh Avenue, Daoli District, Harbin, 150001, Heilongjiang Province, China.
| | - Mingyan Zhao
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, No. 2075, Qunli Seventh Avenue, Daoli District, Harbin, 150001, Heilongjiang Province, China.
| |
Collapse
|
195
|
Xu M, Li X, Yuan C, Zhu T, Wang M, Zhu Y, Duan Y, Yao J, Luo B, Wang Z, Yin S, Zhao Y. Ursolic Acid Inhibits Glycolysis of Ovarian Cancer via KLF5/PI3K/AKT Signaling Pathway. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:2211-2231. [PMID: 39614414 DOI: 10.1142/s0192415x2450085x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2024]
Abstract
Glycolysis is one of the key metabolic reprogramming characteristics of ovarian cancer. Ursolic Acid (UA), as a natural compound, exerts a beneficial regulatory effect on tumor metabolism. In this study, we have confirmed through RNA-seq analysis and a series of in vitro and in vivo functional experiments that UA significantly inhibits ovarian cancer cell proliferation, promotes tumor apoptosis, and reduces glycolysis levels. Additionally, it demonstrates synergistic therapeutic effects with cisplatin in both in vitro and in vivo experiments. Furthermore, at the molecular level, we found that UA inhibits glycolysis in ovarian cancer by binding to the transcription factor KLF5 and blocking the transcriptional expression of the downstream PI3K/AKT signaling pathway, thereby exerting its therapeutic effect. In conclusion, our research indicates that UA can inhibit the proliferation, apoptosis, and glycolysis levels of ovarian cancer cells through the KLF5/PI3K/AKT signaling axis. Our findings offer a new perspective on the therapeutic application of the natural compound UA in ovarian cancer and support its potential development as a candidate for chemotherapy.
Collapse
Affiliation(s)
- Meng Xu
- Cancer Institute, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P. R. China
| | - Xiaoqi Li
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Centre, Shanghai 200032, P. R. China
| | - Chenyue Yuan
- Cancer Institute, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P. R. China
| | - Tingting Zhu
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200090, P. R. China
| | - Mengfei Wang
- Cancer Institute, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P. R. China
| | - Ying Zhu
- Central Laboratory, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P. R. China
| | - Yanqiu Duan
- Central Laboratory, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P. R. China
| | - Jialiang Yao
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P. R. China
| | - Bin Luo
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P. R. China
| | - Ziliang Wang
- Cancer Institute, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P. R. China
| | - Sheng Yin
- Department of Obstetrics and Gynecology, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Yuqing Zhao
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200090, P. R. China
| |
Collapse
|
196
|
He R, Liu Y, Fu W, He X, Liu S, Xiao D, Tao Y. Mechanisms and cross-talk of regulated cell death and their epigenetic modifications in tumor progression. Mol Cancer 2024; 23:267. [PMID: 39614268 PMCID: PMC11606237 DOI: 10.1186/s12943-024-02172-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/07/2024] [Indexed: 12/01/2024] Open
Abstract
Cell death is a fundamental part of life for metazoans. To maintain the balance between cell proliferation and metabolism of human bodies, a certain number of cells need to be removed regularly. Hence, the mechanisms of cell death have been preserved during the evolution of multicellular organisms. Tumorigenesis is closely related with exceptional inhibition of cell death. Mutations or defects in cell death-related genes block the elimination of abnormal cells and enhance the resistance of malignant cells to chemotherapy. Therefore, the investigation of cell death mechanisms enables the development of drugs that directly induce tumor cell death. In the guidelines updated by the Cell Death Nomenclature Committee (NCCD) in 2018, cell death was classified into 12 types according to morphological, biochemical and functional classification, including intrinsic apoptosis, extrinsic apoptosis, mitochondrial permeability transition (MPT)-driven necrosis, necroptosis, ferroptosis, pyroptosis, PARP-1 parthanatos, entotic cell death, NETotic cell death, lysosome-dependent cell death, autophagy-dependent cell death, immunogenic cell death, cellular senescence and mitotic catastrophe. The mechanistic relationships between epigenetic controls and cell death in cancer progression were previously unclear. In this review, we will summarize the mechanisms of cell death pathways and corresponding epigenetic regulations. Also, we will explore the extensive interactions between these pathways and discuss the mechanisms of cell death in epigenetics which bring benefits to tumor therapy.
Collapse
Affiliation(s)
- Ruimin He
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Yifan Liu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Weijie Fu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Xuan He
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Shuang Liu
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Desheng Xiao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Yongguang Tao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China.
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China.
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China.
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- Department of Thoracic Surgery, Hunan Key Laboratory of Early Diagnosis and Precision Therapy in Lung Cancer, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
- Furong Laboratory, Xiangya School of Medicine, Central South University, Hunan, 410078, China.
| |
Collapse
|
197
|
Lin L, Liu H, Zhang D, Du L, Zhang H. Nanolevel Immunomodulators in Sepsis: Novel Roles, Current Perspectives, and Future Directions. Int J Nanomedicine 2024; 19:12529-12556. [PMID: 39606559 PMCID: PMC11600945 DOI: 10.2147/ijn.s496456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
Sepsis represents a profound challenge in critical care, characterized by a severe systemic inflammatory response which can lead to multi-organ failure and death. The intricate pathophysiology of sepsis involves an overwhelming immune reaction that disrupts normal host defense mechanisms, necessitating innovative approaches to modulation. Nanoscale immunomodulators, with their precision targeting and controlled release capabilities, have emerged as a potent solution to recalibrate immune responses in sepsis. This review explores the recent advancements in nanotechnology for sepsis management, emphasizing the integration of nanoparticulate systems to modulate immune function and inflammatory pathways. Discussions detail the development of the immune system, the distinct inflammatory responses triggered by sepsis, and the scientific principles underpinning nanoscale immunomodulation, including specific targeting mechanisms and delivery systems. The review highlights nanoformulation designs aimed at enhancing bioavailability, stability, and therapeutic efficacy, which shows promise in clinical settings by modulating key inflammatory pathways. Ultimately, this review synthesizes the current state of knowledge and projects future directions for research, underscoring the transformative potential of nanolevel immunomodulators for sepsis treatment through innovative technologies and therapeutic strategies.
Collapse
Affiliation(s)
- Liangkang Lin
- Department of Pediatrics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, People’s Republic of China
| | - Hanyou Liu
- Department of Pediatrics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, People’s Republic of China
| | - Dingshan Zhang
- Department of Intensive Care Unit, Public Health Clinical Center of Chengdu, Chengdu, People’s Republic of China
| | - Lijia Du
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, People’s Republic of China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, People’s Republic of China
| | - Haiyang Zhang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, People’s Republic of China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, People’s Republic of China
| |
Collapse
|
198
|
Jiang Y, Song Y, Zeng Q, Jiang B. Mesenchymal Stem Cells and Their Extracellular Vesicles Are a Promising Alternative to Antibiotics for Treating Sepsis. Bioengineering (Basel) 2024; 11:1160. [PMID: 39593820 PMCID: PMC11591657 DOI: 10.3390/bioengineering11111160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/08/2024] [Accepted: 11/10/2024] [Indexed: 11/28/2024] Open
Abstract
Sepsis is a life-threatening disease caused by the overwhelming response to pathogen infections. Currently, treatment options for sepsis are limited to broad-spectrum antibiotics and supportive care. However, the growing resistance of pathogens to common antibiotics complicates treatment efforts. Excessive immune response (i.e., cytokine storm) can persist even after the infection is cleared. This overactive inflammatory response can severely damage multiple organ systems. Given these challenges, managing the excessive immune response is critical in controlling sepsis progression. Therefore, Mesenchymal stem cells (MSCs), with their immunomodulatory and antibacterial properties, have emerged as a promising option for adjunctive therapy in treating sepsis. Moreover, MSCs exhibit a favorable safety profile, as they are eventually eliminated by the host's immune system within several months post-administration, resulting in minimal side effects and have not been linked to common antibiotic therapy drawbacks (i.e., antibiotic resistance). This review explores the potential of MSCs as a personalized therapy for sepsis treatment, clarifying their mechanisms of action and providing up-to-date technological advancements to enhance their protective efficacy for patients suffering from sepsis and its consequences.
Collapse
Affiliation(s)
- Yu Jiang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Chengdu 610041, China
| | - Yunjuan Song
- R&D Division, Eureka Biotech Inc., Philadelphia, PA 19104, USA
| | - Qin Zeng
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Bin Jiang
- R&D Division, Eureka Biotech Inc., Philadelphia, PA 19104, USA
| |
Collapse
|
199
|
Wang C, Zhao X, Wang K, Liang H, Chen S, Liu Y, Yao H, Jiang J. Prospective Application of Mesenchymal Stem Cell-Derived Exosomes in the Treatment of Disseminated Intravascular Coagulation. Int J Nanomedicine 2024; 19:11957-11971. [PMID: 39569063 PMCID: PMC11577934 DOI: 10.2147/ijn.s467158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/05/2024] [Indexed: 11/22/2024] Open
Abstract
Disseminated intravascular coagulation (DIC) is an acquired disorder characterized by systemic activation of blood coagulation, which can arise from various causes. Owing to its abrupt onset, rapid progression, and high mortality rate, DIC presents a major clinical challenge. Anticoagulant drugs, such as heparin or low-molecular-weight heparin, are the current gold standard of treatment; however, these interventions pose considerable bleeding risks. Thus, safer and more effective therapeutic strategies are urgently required. Owing to their strong anti-inflammatory and tissue repair capabilities, mesenchymal stem cell-derived exosomes (MSC-Exos) have gained considerable attention as novel therapeutic options for numerous disorders, including DIC. Their stability in diverse pathological states highlights their potential as promising candidates for DIC therapy. This review presents the latest insights on the pathogenesis of DIC and anti-inflammatory and anticoagulant properties of MSC-Exos. We aimed to elucidate the potential mechanisms by which MSC-Exos influence DIC pathogenesis. We speculate that MSC-Exos offer a multifaceted approach to DIC treatment by attenuating neutrophil extracellular trap formation, modulating M1/M2 macrophage polarization, altering Nrf2/NF-κB signalling pathway to downregulate pro-inflammatory factors, and correcting imbalances in the coagulation-fibrinolysis system through anticoagulant routes. This suggests that MSC-Exos are a potential paradigm in DIC therapy, offering novel targets and treatment modalities for DIC management.
Collapse
Affiliation(s)
- Chengran Wang
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Xiaoqing Zhao
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Keyan Wang
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Huixin Liang
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Shuhan Chen
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Yajie Liu
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Hua Yao
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Jinlan Jiang
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| |
Collapse
|
200
|
Nie Z, Fan Q, Jiang W, Wei S, Luo R, Hu H, Liu G, Lei Y, Xie S. Placental mesenchymal stem cells suppress inflammation and promote M2-like macrophage polarization through the IL-10/STAT3/NLRP3 axis in acute lung injury. Front Immunol 2024; 15:1422355. [PMID: 39620220 PMCID: PMC11604576 DOI: 10.3389/fimmu.2024.1422355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 10/29/2024] [Indexed: 12/23/2024] Open
Abstract
INTRODUCTION Acute lung injury (ALI) is a clinically severe respiratory disorder that currently lacks specific and effective pharmacotherapy. The imbalance of M1/M2 macrophage polarization is pivotal in the initiation and progression of ALI. Shifting macrophage polarization from the proinflammatory M1 phenotype to the anti-inflammatory M2 phenotype could be a potential therapeutic strategy. The intratracheal administration of placental mesenchymal stem cells (pMSCs) has emerged as a novel and effective treatment for ALI. This study aimed to investigate the role and downstream mechanisms of pMSCs in reprogramming macrophage polarization to exert anti-inflammatory effects in ALI. METHODS The study used lipopolysaccharide (LPS) to induce inflammation in both cell and rat models of ALI. Intratracheal administration of pMSCs was tested as a therapeutic intervention. An expression dataset for MSCs cultured with LPS-treated macrophages was collected from the Gene Expression Omnibus database to predict downstream regulatory mechanisms. Experimental validation was conducted through in vitro and in vivo assays to assess pMSCs effects on macrophage polarization and inflammation. RESULTS Both in vitro and in vivo experiments validated that pMSCs promoted M2 macrophage polarization and reduced the release of inflammatory factors. Further analyses revealed that pMSCs activated the signal transducer and activator of transcription (STAT)3 signaling pathway by secreting interleukin (IL)-10, leading to increased STAT3 phosphorylation and nuclear translocation. This activation inhibited NLRP3 inflammasome activation, promoting M2 macrophage polarization and suppressing the inflammatory response. CONCLUSION The study concluded that pMSCs alleviated lung injury in an LPS-induced ALI model by inhibiting M1 macrophage polarization and proinflammatory factor secretion, while promoting M2 macrophage polarization. This effect was mediated via the IL-10/STAT3/NLRP3 axis, presenting a novel therapeutic pathway for ALI treatment.
Collapse
Affiliation(s)
- Zhihao Nie
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qinglu Fan
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wanli Jiang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shujian Wei
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Renwei Luo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Haifeng Hu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Gaoli Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yufei Lei
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Songping Xie
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|